Next Article in Journal
The Potential of T Cell Factor 1 in Sustaining CD8+ T Lymphocyte-Directed Anti-Tumor Immunity
Next Article in Special Issue
Integrative Transcriptomic Network Analysis of Butyrate Treated Colorectal Cancer Cells
Previous Article in Journal
Postoperative Trends and Prognostic Values of Inflammatory and Nutritional Biomarkers after Liver Transplantation for Hepatocellular Carcinoma
Previous Article in Special Issue
Non-Coding RNAs as Mediators of Epigenetic Changes in Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs in the Pathogenesis of Hepatocellular Carcinoma: A Review

Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(3), 514; https://doi.org/10.3390/cancers13030514
Submission received: 21 December 2020 / Revised: 22 January 2021 / Accepted: 25 January 2021 / Published: 29 January 2021
(This article belongs to the Collection Regulatory and Non-Coding RNAs in Cancer Epigenetic Mechanisms)

Abstract

:

Simple Summary

Hepatocellular carcinoma (HCC) is one of the most frequently occurring cancers, and the prognosis for late-stage HCC remains poor. A better understanding of the pathogenesis of HCC is expected to improve outcomes. MicroRNAs (miRNAs) are small, noncoding, single-stranded RNAs that regulate the expression of various target genes, including those in cancer-associated genomic regions or fragile sites in various human cancers. We summarize the central roles of miRNAs in the pathogenesis of HCC and discuss their potential utility as valuable biomarkers and new therapeutic agents for HCC.

Abstract

Hepatocellular carcinoma (HCC) is the seventh most frequent cancer and the fourth leading cause of cancer mortality worldwide. Despite substantial advances in therapeutic strategies, the prognosis of late-stage HCC remains dismal because of the high recurrence rate. A better understanding of the etiology of HCC is therefore necessary to improve outcomes. MicroRNAs (miRNAs) are small, endogenous, noncoding, single-stranded RNAs that modulate the expression of their target genes at the posttranscriptional and translational levels. Aberrant expression of miRNAs has frequently been detected in cancer-associated genomic regions or fragile sites in various human cancers and has been observed in both HCC cells and tissues. The precise patterns of aberrant miRNA expression differ depending on disease etiology, including various causes of hepatocarcinogenesis, such as viral hepatitis, alcoholic liver disease, or nonalcoholic steatohepatitis. However, little is known about the underlying mechanisms and the association of miRNAs with the pathogenesis of HCC of various etiologies. In the present review, we summarize the key mechanisms of miRNAs in the pathogenesis of HCC and emphasize their potential utility as valuable diagnostic and prognostic biomarkers, as well as innovative therapeutic targets, in HCC diagnosis and treatment.

1. Introduction

Liver cancer is the sixth most frequent malignancy and the fourth leading cause of cancer-related deaths worldwide [1]. Hepatocellular carcinoma (HCC) accounts for approximately 80% of all liver cancers and is a main cause of cancer mortality [2]. The risk factors of hepatocarcinogenesis include various liver diseases, such as infections caused by hepatitis B virus (HBV) and hepatitis C virus (HCV), alcoholic liver disease (ALD), nonalcoholic steatohepatitis (NASH), and nonalcoholic fatty liver disease (NAFLD) [3,4,5]. Despite recent great progress in HCC therapy, the 5-year survival rate for advanced-stage HCC remains poor owing to its late diagnosis, resistance to anticancer therapy, and high frequency of recurrence [6,7,8]. Therefore, elucidating the detailed underlying mechanisms and pathogenesis of HCC is important for the development of new diagnostic and prognostic biomarkers and therapeutic drugs.
MicroRNAs (miRNAs, miRs) are small, endogenous, interfering, noncoding RNAs of 21–30 nucleotides in length. More than 2600 miRNAs have been predicted to be encoded by the human genome, with the ability to modulate more than 15,000 genes [9]. Each miRNA negatively regulates target genes by binding to the 3′ untranslated region (UTR) of mRNAs. These complexes are involved in RNA-mediated interference, and in vertebrates, mRNA transcripts are usually not cleaved by a miRNA-associated RNA-induced silencing complex (RISC) but rather undergo translational repression and degradation via deadenylation [10]. The miRNA-associated RISC can suppress gene expression [11,12]. Remarkably, a single miRNA can modulate more than 200 mRNAs [13,14].
The biogenesis of miRNAs includes several steps, such as transcription, cleavage, export, further cleavage, strand selection, and interaction with mRNAs [15] (Figure 1). Generally, RNA polymerase II transcribes primary miRNA (pri-miRNAs) transcripts in the canonical pathway or mirtron pathway [16]. The 5′ ends of pri-miRNAs are capped, and the 3′ ends are polyadenylated [17]. Pri-miRNAs are then cleaved to 70–100-nucleotide hairpin-structured precursors (pre-miRNAs) by a nuclear RNase III enzyme named Drosha, which is a double-stranded RNA-binding domain protein involving DiGeorge syndrome critical region 8 (DGCR8)/Pasha as a cofactor [18]. Next, pre-miRNAs are exported from the nucleus to the cytoplasm, after binding with exportin-5 and Ran-GTP [19] and are cleaved by Dicer [20], with transactivation response element RNA-binding 70 protein (TRBP) serving as a cofactor for Dicer [20]. Finally, double-stranded pre-miRNAs undergo rapid unwinding, when loaded onto Argonaute (AGO), and only one strand, which serves as a guide to target mRNAs, remains bound [21]. These RISCs with the AGO protein modulate mRNA degradation and translational inhibition [21].
However, the biogenesis of miRNAs is more diverse and may involve noncanonical pathways that bypass Drosha/DGCR8 processing. There are several noncanonical types of miRNAs, including mirtrons, small nucleolar RNAs (snoRNAs), transfer RNAs (tRNAs), and short hairpin RNAs (shRNAs) [22]. Mirtrons are pre-miRNAs that are generated without Drosha by pre-mRNA splicing and intron debranching [23,24]. SnoRNAs [25] and tRNAs [26,27] have different internal hairpin structures, and their processing involves the cleavage activity of Dicer, without Drosha/DGCR8. Transcripts of shRNA, which are derived from unannotated and intergenic regions, can function as Dicer substrates during transcription [28,29].
Although the important roles of miRNAs in the modulation of mRNA expression are well established, their precise functions remain elusive. Interestingly, miRNAs modulate the expression of approximately 30% of all human genes, many of which are tumor-associated or are in regions of instability in the genome [30,31]. There is clear evidence of key roles for miRNAs in human carcinogenesis [13,32,33,34,35], with two types of miRNAs identified, namely, oncogenic miRNAs (oncomiRs) and tumor suppressor miRNAs (Figure 1). OncomiRs induce carcinogenesis by inhibiting the expression of tumor suppressors, while tumor suppressor miRNAs inhibit oncogene expression in normal cells and are lacking in cancer cells. Two miRNAs, miR-15 and miR-16, were first reported to be altered in cancer and are associated with a frequently targeted chromosomal deletion of BCL2, which encodes an anti-apoptotic factor [36].
Several reports have demonstrated relationships between miRNAs and HCC [35,37,38,39,40] and identified potential miRNA biomarkers and therapeutic targets for HCC diagnosis and treatment. This review provides a summary of (1) miRNA functions in the liver, (2) associations of miRNAs with the pathogenesis of HCC of various etiologies, and (3) the mechanisms underlying the miRNA effects. Furthermore, we emphasize potential utility of various miRNAs as HCC biomarkers and target molecules for therapeutics.

2. Roles of miRNAs in the Liver

2.1. Lipid Metabolism

The liver plays a critical role in lipid metabolism. Dysfunctions in lipid metabolism induce excessive accumulation of hepatic triglycerides and fatty acids, resulting in various liver diseases, such as NAFLD and NASH. Several miRNAs have pivotal functions in the maintenance of cholesterol and fatty acid metabolism [41], and others, such as miR-33, miR-103, miR-104, and miR-307, act as modulators of lipid and cholesterol levels [42].
Various serum miRNAs, including miR-122, miR-21, miR-34a, and miR-451, are enhanced in patients with NAFLD [43]. A representative liver-specific miRNA, miR-122, which is highly upregulated in the liver, is involved in hepatic cholesterol and lipid metabolism in this disease [44]. Suppression of miR-122 can diminish the plasma cholesterol levels, reduce hepatic fatty acid and cholesterol synthesis, and increase the oxidation of hepatic fatty acids [45]. In addition, miR-122 is associated with hepatic lipogenesis-related enzymes, including fatty acid synthase (FASN) and acetyl-CoA carboxylase (ACC1). The inhibition of miR-122 also decreases hepatic lipogenesis in obese mice [44].
Several reports have demonstrated that miR-34a is associated with hepatic steatosis [46,47]. This miRNA inhibits hepatic silent information regulator 1 (sirtuin 1, SIRT1), peroxisome proliferator-activated receptor-α (PPARα), and liver X receptor (LXR) [46,47,48]. Reduced SIRT1 levels in the liver of patients with NAFLD can be recovered by the inhibition of miR-34a, which results in the improvement of hepatic steatosis via PPARα and the activation of AMP-activated protein kinase (AMPK) [47].

2.2. Glucose Metabolism

miR-375, a novel islet-specific miRNA, can inhibit glucose-induced insulin secretion, whereas inhibition of miR-375 can increase insulin secretion. This miR-375-associated insulin modulation is independent of altered glucose metabolism and is related to insulin exocytosis. Myotrophin (MTPN), which is a target of miR-375, is also involved in glucose metabolism. The inhibition of MTPN enhances glucose-induced insulin secretion and insulin exocytosis, indicating that miR-375 might be a new therapeutic target for diabetes mellitus and NAFLD [49]. Expression of miR-23a is increased in a NASH-related HCC murine model, and overexpression of miR-23a via the interleukin-6 (IL-6)/STAT3 signaling pathway decreases glucose production through the inhibition of PGC1α and G6PC expression [50]. In addition, miR-143, which is involved in insulin resistance, controls the ORP8-dependent regulatory pathway of AKT, and miR-143 overexpression reduces insulin-stimulated AKT activation [51]. Moreover, miR-206 decreases lipid and glucose levels in hepatocytes by modulating lipogenesis and insulin signaling [52], which suggests that miR-206 might be a diagnostic biomarker and therapeutic target for NAFLD and hyperglycemia.

2.3. Hepatic Inflammation

Hepatic inflammation, which involves inflammatory cytokine production and endoplasmic reticulum stress, results from an abnormal immune response; the latter is mediated by several factors, such as viral and bacterial infections, metabolic disorders, alcohol abuse, drug allergies, and toxic reagents [53]. Various miRNAs, such as miR-122 and miR-132, play pivotal roles in the innate and adaptive immunity involved in hepatic inflammation. miR-122 is the most representative miRNA in the liver, and its loss results in inflammation, fibrosis, and HCC, indicating that miR-122 modulates anti-inflammatory effects [54,55]. Furthermore, miR-122 may inhibit hepatic infiltration of inflammatory cells and the secretion of various cytokines, including IL-6 and tumor necrosis factor-α (TNFα), by these cells [54,55]. miR-132, which inhibits the expression of SIRT1, is a mediator of inflammation in chronic liver diseases. The overexpression of miR-132 induces the translocation of nuclear factor-κB (NF-κB) into the nucleus, acetylation of p65, and production of IL-8 and MCP-1. The loss of miR-132, which is mediated by serum deprivation, diminishes the acetylation level of p65 and partially downregulates the expression of IL-8 and MCP-1. Therefore, the inhibition of miR-132 has anti-inflammatory effects in the liver.

2.4. Hepatic Fibrosis

Fibrosis is developed in the liver as a result of continuous and severe hepatocyte damage, resulting in an inflammatory cytokine storm. Several miRNAs can synergistically modulate inflammatory signaling pathways. Furthermore, several miRNAs are associated with the activation of hepatic stellate cells (HSCs) and progression of hepatic fibrosis via the regulation of related signaling pathways.
Members of the miR-29 family induce apoptosis via the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway and modulate the accumulation of the extracellular matrix (ECM) [56,57,58]. Stimulation of HSCs by transforming growth factor-beta (TGF-β) promotes myofibroblastic transition and ECM induction, resulting in liver fibrogenesis. TGF-β1 mediates the downregulation of miR-29 in HSCs [57]. In addition, miR-29 overexpression in mouse HSCs leads to the reduction of collagen-1α1 and collagen-4α1 levels [57,59,60] via modulation of various ECM genes. Similar to other hepatic fibrosis-related miRNAs, members of the miR-34 family promote hepatic fibrosis by activating HSCs, whereas those of the miR-378 family inhibit the development of fibrosis in a GLIS-dependent manner. The miR-15 family is associated with the induction of cell proliferation and apoptosis, and its members regulate the TGF-β signaling pathway by inhibiting TGFβR1, SMAD3, SMAD7, p38, and endoglin in cardiac fibrosis [61]. The miR-199 and miR-200 families are involved in the secretion of profibrotic cytokines and are responsible for ECM deposition [62,63]. These miRNA families act as modulators of hepatic fibrosis by targeting genes involved in fibrosis-related signaling pathways and activating HSCs.

3. Roles of miRNAs in Various Liver Diseases Leading to HCC

3.1. HBV Infection

HBV infection is one of the most prevalent risk factors of HCC development [64]. Thus, HBV-related HCC is a serious health concern worldwide [64]. Recently, many reports have demonstrated that miRNAs play pivotal roles in each stage of HBV-related HCC development [65,66,67,68]. HBV dysregulates miRNAs that modulate the expression of the host/HBV genes during HCC pathogenesis. In fact, this type of HCC is characterized by a range of immune response failures due to the dysregulation of miRNAs [69].
The HBV X protein (HBx) inhibits miR-34 expression via p53 stimulation in hepatocytes, which results in the upregulation of a macrophage-derived chemokine (CCL22), stimulation of regulatory T cells (Tregs), and the suppression of effector T cells, thereby increasing HBV genome transcription [70,71]. HBx-induced upregulation of miR-155 leads to a reduction in the suppressor of cytokine signaling-1 (SOCS1) expression, increasing JAK/STAT signaling and suppressing HBV infection mediated by the induction of interferon (IFN) signaling [72]. In addition, HBx-induced miR-155 blocks CCAAT/enhancer-binding protein (C/EBP), which activates the HBV enhancer (Enh) 11/core promoter and then inhibits HBV replication [73].
Liver-specific miR-122 is upregulated in the serum after HBV infection and is regarded as one of the key modulators of HBV replication [74,75,76]. However, Wang et al. [77] demonstrated that HBx suppressed miR-122 expression and increased the levels of HBV transcripts by blocking p53 binding to the HBV Enh1/core promoter. Another report has revealed that miR-122 blocks HBV pregenome RNA, which encodes the hepatitis B core antigen and viral polymerase and inhibits HBV replication via dysregulation of heme oxygenase-1 (HO-1), which suppresses refill of HBV covalently closed circular DNA [78]. In addition, upregulation of several miRNAs, including miR-184, miR-185, miR-196a, miR-199a-3p, miR-210, and miR-217, directly affects HBV transcription [79]. Furthermore, it has recently been demonstrated that the HBV virion produces its own HBV-miR-2 and HBV-miR-3 [80]. HBV-miR-3 inhibits mRNA expression of the hepatitis B core (HBc) protein, which is involved in HBV self-regulation [81], to prolong survival by escaping the host immune system [79,82]. miR-372 also modulates HBV expression, which depends on target pathways. HBx-upregulated miR-372 [83,84] targets the cyclic AMP response element-binding protein (CREB) by binding to HBV Enh1/core promoter (ENI-Cp), thereby suppressing HBV transcription. miR-372 targets nuclear factor 1 B-type protein (NFIB), which modulates HBV Enh1/ENI-Cp, thus increasing HBV transcript levels [79,83]. Various miRNAs are thus related to the modulation of HBV transcript levels and the host immune response to the virus.

3.2. HCV Infection

HCV contributes to serious liver problems, such as liver cancer, including HCC [85]. IFN-free, direct-acting antiviral therapy improves HCV treatment, with sustained virologic response rates greater than 95%, without many side effects [86]. However, the high mutation rate of HCV may result in the resistance to direct-acting antivirals, and patients with mutated HCV have low sustained virologic response rates [87]. Therefore, further studies of the molecular mechanism underlying HCV infections are needed to improve HCV therapy.
Dysregulation of miRNAs due to HCV infection occurs via multiple pathways, such as the immune response, lipid metabolism, and cell-cycle pathways [88]. Several target genes, such as PPARG and fibronectin 1 (FN1), were found to be downregulated, while other target genes, including stearoyl-CoA desaturase (SCD) and CREB1, were found to be upregulated by at least 11 miRNAs, namely, miR-130a/b, miR-200, miR-34a, miR-23b, miR-24, miR-146a, miR-381, miR-25, miR-200a, and miR-371-5p, after HCV infection [89]. In addition, miR-122 modulates the host immune response by regulating the expression of target genes and directly targeting the HCV genome [90,91,92]. In particular, miR-122 stabilizes the 5′ and 3′ UTRs of the HCV genome, and inhibition of this miRNA dramatically reduces the replication of HCV RNA [93,94]. By contrast, miR-141, miR-192, miR-215, and miR-491 increase HCV replication. Thus, miR-141 enhances HCV replication by reducing the tumor suppressor deleted in liver cancer 1 (DLC-1) [95], and miR-491 enables HCV entry via the PI3K/AKT signaling pathway [96]. However, miR-196, miR-29, let-7b, miR-130a, and miR-27a induce anti-HCV activity [59,97,98,99,100,101]. It has also been found that miR-199a suppresses HCV replication by blocking domain II of the internal ribosomal entry site [97]. In addition, IFN-β enhances the expression of several miRNAs with anti-HCV activity, such as miR-196, miR-431, and miR-448. Among these, miR-196 and miR-448 may directly target the HCV RNA genome [91]. These findings suggest that several miRNAs are strongly involved in the modulation of HCV infection and replication.

3.3. ALD

ALD is a complex disease caused by prolonged and heavy alcohol consumption, along with predisposing genetic factors. ALD can cause liver dysfunction, including steatohepatitis, fibrosis, cirrhosis, and eventually HCC [102]. There is tremendous evidence of the effects of miRNA dysregulation on the pathogenesis and development of ALD, such as liver damage, lipid metabolism dysfunction, inflammation, oxidative stress, apoptosis, and fibrosis [102]. Inflammation-related miRNAs, such as miR-132, miR-155, miR-146, and miR-21, influence alcohol/lipopolysaccharide (LPS)/TLR4 pathways. TLR4 transmits proinflammatory stimuli via a mitogen-activated protein kinases (MAPKs) or TIR domain-containing adaptor-inducing IFN-β (TRIF) [103]. The expression of miR-212 is increased by alcohol in intestinal cells, and LPS is also induced by alcohol in the liver via the suppression of ZO-1 [104,105]. Alcohol-induced oxidative stress downregulates miR-199a in liver sinusoidal endothelial cells by upregulating ET1 and hypoxia-inducible factor 1α (HIF1α), which are related to steatohepatitis and fibrosis in the liver [106]. The downregulation of miR-223 increases neutrophil infiltration in the liver and induces liver injury by inhibiting the IL-6/p47phox/reactive oxygen species axis [107]. The enhanced expression of miR-217 exacerbates alcoholic fatty changes by damaging SIRT1 and lipin-1 [108]. Moreover, upregulation of miR-34a in ALD results in the development of liver fibrosis via caspase-2, SIRT1, and matrix metallopeptidase (MMP) 1 and MMP2 [109]. In addition, miR-122 modulates hepatic lipid metabolism and inflammation [110]. The miRNA let-7 is diminished in response to alcohol and loss of let-7 induces a mesenchymal phenotype in HSCs to enhance liver injury by inhibiting LIN28B. Finally, an imbalance between let-7 and LIN28/28B causes oncogenesis [111].

3.4. NAFLD and NASH

NAFLD is defined as steatosis in at least 5% of hepatocytes [112], without other liver diseases, including viral hepatitis and autoimmune, alcohol-related, and genetic liver diseases. Recently, NAFLD has become one of the most frequent liver diseases worldwide [113]. Various miRNAs have been identified to be involved in NASH development. For example, the levels of miR-122, which is a representative hepatic miRNA, are 7.2 times higher in patients with NASH than in healthy subjects and 3.1 times higher than in patients with simple steatosis [114]. Liver-specific miR-122-knockout mice rapidly develop NASH and exhibit enhanced lipogenesis, changes in lipid secretion, IL-6 and TNF-α production, and upregulation of chemokine (C–C motif) ligand 2 (CCL2). Decreased miR-122 expression enhances fibrogenesis by inducing HIF1α and MAPK1, which can also facilitate HCC development [115]. In addition, miR-192 is involved in the development of TGF-β1-promoted fibrosis, which activates SMAD signaling [116]. However, members of a miRNA superfamily that includes miR-16, miR-497, miR-195, miR-322, and miR-15, particularly miR-15 and miR-16, regulate hepatic fibrosis and hepatocarcinogenesis [61]. The development and progression of NASH increases the risk of HCC via miRNAs. A recent report has demonstrated that miRNAs are pivotal for the activation of HSCs during NASH development [117]. Free cholesterol is accumulated because of increases in both SREBP2 and miR-33a signaling via the inhibition of PPARγ signaling, along with HSC activation and disruption of the SREBP2-induced cholesterol feedback system [117]. Upregulation of miR-21, which downregulates the expression of the tumor suppressor phosphatase and tensin homolog (PTEN), is mediated by unsaturated fatty acids in hepatocytes [118]. Furthermore, the expression of miR-155, which inhibits another tumor suppressor gene, C/EBPβ, is enhanced in mice fed a choline-deficient, amino acid-defined diet [73,119].

4. Dysregulation of miRNAs and HCC Development

4.1. miRNAs Involved in Carcinogenesis

Dysregulation of miRNAs is observed in various cancer types [120,121,122,123,124,125]. Although several tumor suppressor miRNAs, which target oncogenes, are downregulated, other miRNAs (oncomiRs), which target tumor suppressor genes, are upregulated in HCC. Genomic regions encoding miRNAs associated with dysfunction can protect against genetic mutations. Carcinogenesis-related transcription factors, such as MYC, suppress some miRNAs, whereas epigenetic regulation of other miRNAs occurs via DNA methylation and histone modifications [126]. In addition, the loss of miRNA processing machinery genes, such as DROSHA, DGCR8, DICER1, TRBP, and AGO2, reduces mature miRNA synthesis and results in hepatocarcinogenesis and HCC development [126]. Various HCC-associated miRNAs, including miR-21, miR-221, and miR-222, are increased in HCC [35,38,39], whereas others, such as miR-122a, miR-145, miR-199a, and miR-223, are decreased in this disease [37,68]. Several HCC-specific miRNAs have recently been classified (Table 1). In our previous study, we demonstrated that miRNA profiles differed between hepatocytes and HCC cells [127]. This finding suggests that the study of miRNA profiles during HCC development and progression can provide valuable biomarkers and therapeutic targets.

4.2. miRNAs Associated with HCC Development

miRNAs have pivotal functions in cancer development [286], and their profiles differ between normal tissues and various cancers. Many reports have demonstrated that the ectopic expression of miRNAs has oncogenic or tumor-suppressive effects on HCC [287]. Therefore, these miRNAs might be targets for HCC therapy.

4.2.1. Upregulated miRNAs Involved in HCC Development (Oncogenic miRNAs)

miR-21, an oncogenic miRNA, is a potential diagnostic biomarker for HCC and is frequently overexpressed in various cancers [15,288]. In addition to HCC, it is upregulated in several liver diseases, such as viral hepatitis, NAFLD, ALD, and liver fibrosis [289]. miR-21 promotes fibrosis by activating HSCs, which results in hepatocarcinogenesis [290,291,292,293]. In patients with HCC, the expression levels of miR-21 are significantly upregulated in both serum and tissues [289,294] and are correlated with tumor development [295]. Additionally, enhanced miR-21 expression promotes cancer cell migration and invasion by inhibiting Krüppel-like factor 5 (KLF5) in vitro [296]. Chen et al. [297] demonstrated that secreted exosomal miR-21 activated the pyruvate dehydrogenase kinase 1 (PDK1)/AKT pathway in HSCs located near HCC (cancer-associated fibroblasts) by directly targeting PTEN and promoted cancer progression by inducing the secretion of angiogenic molecules, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), MMP2, MMP9, and TGF-β by HCC cells.
miR-155 is an inflammation-related miRNA in various liver diseases, including hepatitis B and NASH [298,299,300], which enhances liver carcinogenesis [301]. miR-155 plays important roles as an oncomiR by inhibiting various tumor suppressors, such as SOCS1 [302], tumor protein p53-inducible nuclear protein 1 (TP53INP1) [303], and mutL homolog 1 (MLH1) [304]. In addition, miR-155 overexpression enhances cell proliferation via activation of the Wnt/β-catenin [305], AKT [306], and JAK2/STAT3 [307] pathways. However, Chen et al. [308] demonstrated that in human cancer, miR-155 could act as a tumor suppressor by modulating claudin-1 (CLDN1) and SMAD2 and as an oncomiR under different circumstances.
miR-221 and miR-222 are the most highly deregulated miRNAs in HCC tissues [260,309]. miR-221 targets key tumor suppressors, including CDKN1B/p27 [310,311,312], CDKN1C/p57 [313,314], PTEN [315], TIMP3 [315], and DNA damage-inducible transcript 4 (DDIT4) [260]. Callegari et al. [257] developed transgenic models overexpressing miR-221/222 in the liver and confirmed that dysregulation of these miRNAs led to hepatocarcinogenesis. These studies suggest that miR-221 is a key oncogenic miRNA in HCC.

4.2.2. Downregulated miRNAs Involved in HCC Development (Tumor Suppressors)

Tumor suppressor miRNAs, including the let-7 family, target the RAS family [288,316]. Members of the let-7 family exert antifibrotic effects at the transcriptional level via PPARγ. These miRNAs regulate various target genes associated with liver fibrosis, thereby diminishing tumorigenesis [317,318]. Remarkably, expression of the members of the let-7 family is diminished in HCV-associated HCC [136]. Among let-7 family members, let-7g levels are notably lower in metastatic HCC than in HCC without metastasis, while higher expression of let-7g in HCC tissues predicts a better prognosis in patients with HCC [136].
Members of the miR-15 family have a common seed sequence and inhibit specific mRNAs. In HCC tissues, members of this family are diminished as tumor suppressors and modulate inflammation by inhibiting IκB kinase (IKKα) and TGF-β-activated kinase 1 binding protein 3 (TAB3) [319]. miR-195, which is in the miR-15 family, suppresses angiogenesis by targeting VEGF, Vav guanine nucleotide exchange factor 2 (VAV2), and cell division cycle 42 (CDC42) in HCC cell lines [320]. A recent report has also demonstrated that miR-497, which also belongs to the miR-15 family, modulates the protein kinase B (PKB) pathway by targeting RICTOR in the HCC cell lines and inhibits cell growth, invasion, and metastasis via the RICTOR/AKT signaling pathway [222].
miR-29 acts as a tumor suppressor in several cancers and regulates oncogenic processes, including epigenetic modifications, proliferation, apoptosis, fibrosis, and angiogenesis [321]. Overexpression of miR-29 induces apoptosis and inhibits tumorigenesis by directly inhibiting anti-apoptotic proteins, including BCL2 and MCL1, based on an in vivo study of HCC [322]. Loss of miR-101, which targets MCL1, induces apoptosis and inhibits tumorigenesis [158]. Furthermore, miR-101 targets Rho-associated protein kinase (ROCK), which is a downstream effector of RhoA GTPase and modulates actomyosin bundles and focal adhesions involved in the inhibition of epithelial–mesenchymal transition (EMT). The overexpression of ROCK2, which is frequently detected in HCC tissues, induces cancer cell motility and invasiveness [323].
miR-122, which is a representative miRNA in the liver, plays a critical role in lipid metabolism and HCV replication, as well as in hepatocarcinogenesis. Thus, miR-122 directly inhibits cyclin G1, IGF-1R, ADAM10, and pyruvate kinase M2 (PKM2) as a tumor suppressor in HCC [324]. Interestingly, in miR-122-knockout mice, the loss of miR-122 causes steatohepatitis and carcinogenesis in the liver [54]. Furthermore, restoration of miR-122 prevents hepatocarcinogenesis in a mouse model [324].

4.2.3. miRNAs and the Tumor Microenvironment (Angiogenesis and Immune Modulation)

HCC is a hypervascular tumor, and angiogenesis is one of the most important factors during HCC development, including proliferation, invasion, and metastasis of the tumor. VEGFA is a critical mitogen for endothelial cells and plays an important role in tumor vessel formation [325]. Several studies have demonstrated that the expression of VEGFA is upregulated in HCC and is associated with oncogenesis [326,327]. Some miRNAs, such as miR-15b, miR-125b, miR-423-3p, miR-424, miR-494, miR-497, miR-612, miR-637, and miR-1225b, are involved in angiogenesis, including modulation of VEGF expression [328].
Recent reports have demonstrated that some specific miRNAs could play important roles in the tumor microenvironment during HCC development [71]. TGF-β inhibits miR-34a expression, resulting in chemokine CCL22 upregulation, which increases the recruitment of Tregs to facilitate immune escape [71]. In addition, Tregs exhibit altered expression of several specific miRNAs, including miR-182-5p, miR-214-3p, miR-129-5p, and miR-30b-5p, and are influenced by FOXP3 in HCC [329]. Li et al. [330] also demonstrated that miR-125b inhibited Treg accumulation and functions induced by Ganoderma lucidum polysaccharide extract, resulting in the suppression of HCC growth. These findings suggest that the expression of specific miRNAs is associated with tumor growth, not only from cancer cells but also from stromal cells, including endothelial and immune cells, during HCC development.

4.3. miRNAs as Biomarkers for HCC

The prognosis of HCC remains poor, as many patients are diagnosed at an advanced stage. Tumor markers, such as α-fetoprotein (AFP) and protein induced by vitamin K absence or antagonists-II (PIVKA-II), are widely used for HCC screening. However, the diagnosis of HCC using these tumor markers has yielded unsatisfactory results [331]. Therefore, new effective biomarkers are required, especially those involved in early HCC.
Many studies have demonstrated the diagnostic and predictive value of several miRNAs in HCC (Table 2). Circulating miRNAs are stably detected in the blood [332] and are therefore one of the most valuable biomarkers for HCC. Basically, miRNAs secreted from cancer cells are contained in exosomes or apoptotic bodies or are bound to serum proteins or lipids [333]. Among miRNAs that are altered in HCC, the downregulated miRNAs, including let-7g [136], miR-22 [334], miR-26 [335], miR-29 [322], miR-99a [156], miR-122 [336], miR-124 [337], miR-139 [338], miR-145 [182], and miR-199b [339], are associated with cell growth, apoptosis, carcinogenesis, angiogenesis, migration [340], invasion, metastasis, and EMT (Table 2). Upregulated miRNAs, such as miR-10b [233], miR-17-5p [341], miR-21 [342], miR-135a [242], miR-155 [343], miR-182 [249], miR-221 [342], and miR-222 [173,342], are associated with metastasis, angiogenesis, and a poor prognosis (Table 2). Additionally, miR-500 is upregulated in the serum of patients with HCC and is downregulated after surgical resection of the tumor [344]. Moreover, miR-25, miR-375, and let-7f can also be used to distinguish patients with HCC from normal controls [345]. These findings indicate that miRNAs can be promising new biomarkers for HCC.

4.4. miRNAs as Therapeutic Targets for HCC

Various miRNAs have been demonstrated to modulate multiple targets in HCC-associated signaling pathways, and miRNA-targeted cancer treatment is an attractive approach for the prevention of HCC development. Recently, reduced miRNA expression in HCC has been found to be restored with replacement therapy, leading to the inhibition of transcription of critical mRNAs that suppress cell proliferation or induce apoptosis in HCC cell lines [353]. By contrast, enhanced expression of miRNAs can be inactivated by antagonists in HCC cells [354]. However, the administration of miRNAs has not resulted in sufficient gene knockdown, since cell membrane permeability is poor, and the nucleus is degraded. To overcome these obstacles, miRNA oligomers, including anti-miRNA oligonucleotides, locked nucleic acid antisense oligonucleotides, miRNA sponges, miRNA masks, nanoparticles, antagomirs, and multiple-target anti-miRNA antisense oligodeoxyribonucleotides, have recently been used to modulate miRNAs [355,356]. Targeting of candidate miRNAs has been developed as a new approach for HCC therapy.
Adeno-associated virus (AAV) vectors can be utilized for the delivery of miRNA inhibitors or miRNA precursors. Notably, miR-26a, which is expressed at low levels in HCC tissues and at high levels in the normal liver, suppresses HCC development when delivered using AAV vectors. In addition, miR-26a replacement induces cell-cycle arrest by inhibiting cyclins D2 and E2 in HCC cells [146]. Several potential targets, including miR-26a [147], miR-122 [357], and miR-124 [164], and promising strategies have been reported for miRNA replacement therapies. The inhibition of miR-221 results in a longer survival, reduction in nodule numbers, and retardation of tumor development [257,358]. Moreover, no toxicity was observed when miRNAs were used for the treatment of HCC in a mouse model. Interestingly, miRNAs influence the sensitivity of various cancers to anticancer drugs. Several studies have demonstrated that the patterns of miRNA expression are significantly changed after metformin treatment, in various cancers, such as gastric cancer [359], esophageal cancer [360], and HCC [361]. Notably, the enhanced expression of miR-21 [362] and miR-181b [248] induces chemoresistance to IFN–5-fluorouracil and doxorubicin therapy in HCC. By contrast, restoration of miR-122 as a tumor suppressor confers sensitivity to sorafenib to HCC cell lines via the downregulation of multidrug resistance genes.

4.5. miRNAs as Prognostic Markers for HCC

As shown in Table 2, recent studies have demonstrated that miRNAs, including miR-92a, miR-221, miR-487a, and miR-1468, have the potential to serve as prognostic biomarkers for HCC, and upregulation of these miRNAs indicates a poor prognosis for patients with HCC [113,126,131,206]. However, downregulation of miRNAs such as miR-33a, miR-137, miR-194, or miR-940 is also associated with a poor prognosis in HCC [143,149,175,207]. In addition, overexpression of exosomal miR-32-5p, which inhibits PTEN [208], and a low expression level of exosomal miR-638 [209] were found to be associated with a poor prognosis in patients with HCC. Furthermore, the expression levels of miR-122, miR-148a, and miR-1246 were found to be significantly higher in serum exosomes from patients with HCC than in those from patients with liver cirrhosis and from healthy controls [210].

5. Conclusions

Our understanding of biological functions of miRNAs and their diagnostic and therapeutic value in HCC is rapidly increasing. Various miRNAs have been identified to be involved in the modulation of biological processes during HCC development. In this review, we provided an overview of the roles of miRNAs in the development and progression of HCC. The promising findings for miRNA applications as HCC biomarkers and therapeutic targets indicate that (i) circulating miRNAs are stable in serum exosomes of patients with HCC and (ii) miRNAs can regulate multiple target genes and signaling pathways in HCC. Therefore, a combination of conventional molecular targeting agents and miRNA-based therapies for HCC might improve the efficiency of transgene expression and gene transfer in primary and metastatic HCC. Although miRNAs are potential biological target molecules for HCC diagnosis and therapy, the number of miRNA-based clinical trials is still insufficient. Major obstacles of the clinical application of miRNAs include inefficient drug delivery, off-target effects, and difficulties in the determination of efficacious doses. Site-specific delivery of miRNAs to HCC tissues and efficacious doses might minimize the off-target effects and toxicity. The precise targetable miRNAs and the mechanisms underlying the miRNA-induced anticancer effects in vivo remain unclear. However, innovative applications are increasing in various fields. Further analyses and new technologies for miRNAs will provide novel insights into the pathogenesis of HCC. Therefore, global miRNA analyses are expected to provide a basis for HCC therapy.

Author Contributions

Conceptualization, A.M.; methodology, A.M.; formal analysis, A.M., K.O., T.T., and T.M.; investigation, A.M., K.O., T.T., and T.M.; writing—original draft preparation, A.M.; writing—review and editing, K.F., J.T., and T.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Aly, A.; Ronnebaum, S.M.; Patel, D.; Doleh, Y.; Benavente, F. Epidemiologic, humanistic and economic burden of hepatocellular carcinoma in the USA: A systematic literature review. Hepatic Oncol. 2020, 7, HEP27. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fujiwara, N.; Friedman, S.L.; Goossens, N.; Hoshida, Y. Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J. Hepatol. 2018, 68, 526–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef]
  5. Younossi, Z.M.; Blissett, D.; Blissett, R.; Henry, L.; Stepanova, M.; Younossi, Y.; Racila, A.; Hunt, S.; Beckerman, R. The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe. Hepatology 2016, 64, 1577–1586. [Google Scholar] [CrossRef]
  6. Altekruse, S.F.; Henley, S.J.; Cucinelli, J.E.; McGlynn, K.A. Changing Hepatocellular Carcinoma Incidence and Liver Cancer Mortality Rates in the United States. Am. J. Gastroenterol. 2014, 109, 542–553. [Google Scholar] [CrossRef] [Green Version]
  7. Zhang, G.; Li, R.; Deng, Y.; Zhao, L. Conditional survival of patients with hepatocellular carcinoma: Results from the Surveillance, Epidemiology, and End Results registry. Expert Rev. Gastroenterol. Hepatol. 2018, 12, 515–523. [Google Scholar] [CrossRef]
  8. Xu, L.; Kim, Y.; Spolverato, G.; Gani, F.; Pawlik, T.M. Racial disparities in treatment and survival of patients with hepatocellular carcinoma in the United States. Hepatobiliary Surg. Nutr. 2016, 5, 43–52. [Google Scholar] [CrossRef]
  9. Chou, C.-H.; Shrestha, S.; Yang, C.-D.; Chang, N.-W.; Lin, Y.-L.; Liao, K.-W.; Huang, W.-C.; Sun, T.-H.; Tu, S.-J.; Lee, W.-H.; et al. miRTarBase update 2018: A resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018, 46, D296–D302. [Google Scholar] [CrossRef]
  10. Suzuki, H.; Maruyama, R.; Yamamoto, E.; Kai, M. Epigenetic alteration and microRNA dysregulation in cancer. Front. Genet. 2013, 4, 258. [Google Scholar] [CrossRef] [Green Version]
  11. Davis-Dusenbery, B.N.; Hata, A. MicroRNA in Cancer: The Involvement of Aberrant MicroRNA Biogenesis Regulatory Pathways. Genes Cancer 2010, 1, 1100–1114. [Google Scholar] [CrossRef] [PubMed]
  12. Farazi, T.A.; Hoell, J.I.; Morozov, P.; Tuschl, T. MicroRNAs in Human Cancer. Adv. Exp. Med. Bio. 2013, 774, 1–20. [Google Scholar] [CrossRef] [Green Version]
  13. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  14. Krek, A.; Grün, D.; Poy, M.N.; Wolf, R.; Rosenberg, L.; Epstein, E.J.; MacMenamin, P.; Da Piedade, I.; Gunsalus, K.C.; Stoffel, M.; et al. Combinatorial microRNA target predictions. Nat. Genet. 2005, 37, 495–500. [Google Scholar] [CrossRef]
  15. Wang, X.W.; Heegaard, N.H.H.; Orum, H. MicroRNAs in Liver Disease. Gastroenterology 2012, 142, 1431–1443. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, V.N.; Han, J.; Siomi, M.C. Biogenesis of small RNAs in animals. Nat. Rev. Mol. Cell Biol. 2009, 10, 126–139. [Google Scholar] [CrossRef] [PubMed]
  17. Cai, X.; Hagedorn, C.H.; Cullen, B.R. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA 2004, 10, 1957–1966. [Google Scholar] [CrossRef] [Green Version]
  18. Gregory, R.I.; Yan, K.-P.; Amuthan, G.; Chendrimada, T.; Doratotaj, B.; Cooch, N.; Shiekhattar, R. The Microprocessor complex mediates the genesis of microRNAs. Nature 2004, 432, 235–240. [Google Scholar] [CrossRef]
  19. Okada, C.; Yamashita, E.; Lee, S.J.; Shibata, S.; Katahira, J.; Nakagawa, A.; Yoneda, Y.; Tsukihara, T. A High-Resolution Structure of the Pre-microRNA Nuclear Export Machinery. Science 2009, 326, 1275–1279. [Google Scholar] [CrossRef]
  20. Chendrimada, T.P.; Gregory, R.I.; Kumaraswamy, E.; Norman, J.; Cooch, N.; Nishikura, K.; Shiekhattar, R. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature 2005, 436, 740–744. [Google Scholar] [CrossRef]
  21. Kim, Y.; Kim, V.N. MicroRNA Factory: RISC Assembly from Precursor MicroRNAs. Mol. Cell 2012, 46, 384–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Xia, J.; Zhang, W. Noncanonical MicroRNAs and Endogenous siRNAs in Lytic Infection of Murine Gammaherpesvirus. PLoS ONE 2012, 7, e47863. [Google Scholar] [CrossRef] [PubMed]
  23. Okamura, K.; Hagen, J.W.; Duan, H.; Tyler, D.M.; Lai, E.C. The Mirtron Pathway Generates microRNA-Class Regulatory RNAs in Drosophila. Cell 2007, 130, 89–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ruby, J.G.; Jan, C.H.; Bartel, D.P. Intronic microRNA precursors that bypass Drosha processing. Nature 2007, 448, 83–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ender, C.; Krek, A.; Friedländer, M.R.; Beitzinger, M.; Weinmann, L.; Chen, W.; Pfeffer, S.; Rajewsky, N.; Meister, G. A Human snoRNA with MicroRNA-Like Functions. Mol. Cell 2008, 32, 519–528. [Google Scholar] [CrossRef] [PubMed]
  26. Blelloch, R. Small RNAs—Their biogenesis, regulation and function in embryonic stem cells. StemBook 2008. [Google Scholar] [CrossRef]
  27. Babiarz, J.E.; Ruby, J.G.; Wang, Y.; Bartel, D.P.; Blelloch, R. Mouse ES cells express endogenous shRNAs, siRNAs, and other Microprocessor-independent, Dicer-dependent small RNAs. Genes Dev. 2008, 22, 2773–2785. [Google Scholar] [CrossRef] [Green Version]
  28. Babiarz, J.E.; Hsu, R.; Melton, C.; Thomas, M.; Ullian, E.M.; Blelloch, R. A role for noncanonical microRNAs in the mammalian brain revealed by phenotypic differences in Dgcr8 versus Dicer1 knockouts and small RNA sequencing. RNA 2011, 17, 1489–1501. [Google Scholar] [CrossRef] [Green Version]
  29. Chiang, H.R.; Schoenfeld, L.W.; Ruby, J.G.; Auyeung, V.C.; Spies, N.; Baek, D.; Johnston, W.K.; Russ, C.; Luo, S.; Babiarz, J.E.; et al. Mammalian microRNAs: Experimental evaluation of novel and previously annotated genes. Genes Dev. 2010, 24, 992–1009. [Google Scholar] [CrossRef] [Green Version]
  30. Garzon, R.; Fabbri, M.; Cimmino, A.; Calin, G.A.; Croce, C.M. MicroRNA expression and function in cancer. Trends Mol. Med. 2006, 12, 580–587. [Google Scholar] [CrossRef]
  31. Si, W.; Shen, J.; Zheng, H.; Fan, W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin. Epigenetics 2019, 11, 1–24. [Google Scholar] [CrossRef] [PubMed]
  32. Michael, M.Z.; Connor, S.M.O.; Pellekaan, N.G.V.H.; Young, G.P.; James, R.J. Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol. Cancer Res. 2003, 1, 882–891. [Google Scholar] [PubMed]
  33. Lee, E.J.; Gusev, Y.; Jiang, J.; Nuovo, G.J.; Lerner, M.R.; Frankel, W.L.; Morgan, D.L.; Postier, R.G.; Brackett, D.J.; Schmittgen, T.D. Expression profiling identifies microRNA signature in pancreatic cancer. Int. J. Cancer 2007, 120, 1046–1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Takahashi, T.; Konishi, H.; Yanagisawa, K.; Tomida, S.; Osada, H.; Endoh, H.; Harano, T.; Yatabe, Y.; Nagino, M.; Nimura, Y.; et al. Reduced Expression of the let-7 MicroRNAs in Human Lung Cancers in Association with Shortened Postoperative Survival. Cancer Res. 2004, 64, 3753–3756. [Google Scholar] [CrossRef] [Green Version]
  35. Meng, F.; Henson, R.; Wehbe–Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 Regulates Expression of the PTEN Tumor Suppressor Gene in Human Hepatocellular Cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [Green Version]
  36. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Nonlinear partial differential equations and applications: Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [Green Version]
  37. Gramantieri, L.; Ferracin, M.; Fornari, F.; Veronese, A.; Sabbioni, S.; Liu, C.-G.; Calin, G.A.; Giovannini, C.; Ferrazzi, E.; Grazi, G.L.; et al. Cyclin G1 Is a Target of miR-122a, a MicroRNA Frequently Down-regulated in Human Hepatocellular Carcinoma. Cancer Res. 2007, 67, 6092–6099. [Google Scholar] [CrossRef] [Green Version]
  38. Wong, Q.W.; Lung, R.W.; Law, P.T.; Lai, P.B.; Chan, K.Y.; To, K.; Wong, N. MicroRNA-223 Is Commonly Repressed in Hepatocellular Carcinoma and Potentiates Expression of Stathmin1. Gastroenterology 2008, 135, 257–269. [Google Scholar] [CrossRef]
  39. Varnholt, H.; Drebber, U.; Schulze, F.; Wedemeyer, I.; Schirmacher, P.; Dienes, H.-P.; Odenthal, M. MicroRNA gene expression profile of hepatitis C virus-associated hepatocellular carcinoma. Hepatology 2008, 47, 1223–1232. [Google Scholar] [CrossRef]
  40. Huang, X.-H.; Wang, Q.; Chen, J.-S.; Fu, X.-H.; Chen, X.-L.; Chen, L.-Z.; Li, W.; Bi, J.; Zhang, L.-J.; Fu, Q.; et al. Bead-based microarray analysis of microRNA expression in hepatocellular carcinoma: miR-338 is downregulated. Hepatol. Res. 2009, 39, 786–794. [Google Scholar] [CrossRef]
  41. Yu, G.; Yang, Z.; Peng, T.; Lv, Y. Circular RNAs: Rising stars in lipid metabolism and lipid disorders. J. Cell. Physiol. 2020. [Google Scholar] [CrossRef]
  42. Rottiers, V.; Näär, A.M. MicroRNAs in metabolism and metabolic disorders. Nat. Rev. Mol. Cell Biol. 2012, 13, 239–250. [Google Scholar] [CrossRef]
  43. Yamada, H.; Suzuki, K.; Ichino, N.; Ando, Y.; Sawada, A.; Osakabe, K.; Sugimoto, K.; Ohashi, K.; Teradaira, R.; Inoue, T.; et al. Associations between circulating microRNAs (miR-21, miR-34a, miR-122 and miR-451) and non-alcoholic fatty liver. Clin. Chim. Acta 2013, 424, 99–103. [Google Scholar] [CrossRef] [PubMed]
  44. Cheung, O.; Puri, P.; Eicken, C.; Contos, M.J.; Mirshahi, F.; Maher, J.W.; Kellum, J.M.; Min, H.; Luketic, V.A.; Sanyal, A.J. Nonalcoholic steatohepatitis is associated with altered hepatic MicroRNA expression. Hepatology 2008, 48, 1810–1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Clarke, J.; Sharapova, T.; Lake, A.D.; Blomme, E.; Maher, J.; Cherrington, N.J. Circulating microRNA 122 in the methionine and choline-deficient mouse model of non-alcoholic steatohepatitis. J. Appl. Toxicol. 2014, 34, 726–732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Salvoza, N.C.; Klinzing, D.C.; Gopez-Cervantes, J.; Baclig, M.O. Association of Circulating Serum miR-34a and miR-122 with Dyslipidemia among Patients with Non-Alcoholic Fatty Liver Disease. PLoS ONE 2016, 11, e0153497. [Google Scholar] [CrossRef] [PubMed]
  47. Ding, J.; Li, M.; Wan, X.; Jin, X.; Chen, S.; Yu, C.; Li, Y. Effect of miR-34a in regulating steatosis by targeting PPARα expression in nonalcoholic fatty liver disease. Sci. Rep. 2015, 5, 13729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Braza-Boïls, A.; Marí-Alexandre, J.; Molina, P.; Arnau, M.A.; Barceló-Molina, M.; Domingo, D.; Girbes, J.; Giner, J.; Martínez-Dolz, L.; Zorio, E. Deregulated hepatic microRNAs underlie the association between non-alcoholic fatty liver disease and coronary artery disease. Liver Int. 2016, 36, 1221–1229. [Google Scholar] [CrossRef]
  49. Poy, M.N.; Eliasson, L.; Krutzfeldt, J.; Kuwajima, S.; Ma, X.; Macdonald, P.E.; Pfeffer, S.; Tuschl, T.; Rajewsky, N.; Rorsman, P.; et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004, 432, 226–230. [Google Scholar] [CrossRef]
  50. Wang, B.; Hsu, S.-H.; Frankel, W.; Ghoshal, K.; Jacob, S.T. Stat3-mediated activation of microRNA-23a suppresses gluconeogenesis in hepatocellular carcinoma by down-regulating Glucose-6-phosphatase and peroxisome proliferator-activated receptor gamma, coactivator 1 alpha. Hepatology 2012, 56, 186–197. [Google Scholar] [CrossRef] [Green Version]
  51. Jordan, S.D.; Krüger, M.; Willmes, D.M.; Redemann, N.; Wunderlich, F.T.; Brönneke, H.S.; Merkwirth, C.; Kashkar, H.; Olkkonen, V.M.; Böttger, T.; et al. Obesity-induced overexpression of miRNA-143 inhibits insulin-stimulated AKT activation and impairs glucose metabolism. Nature 2011, 13, 434–446. [Google Scholar] [CrossRef] [PubMed]
  52. Wu, H.; Zhang, T.; Pan, F.; Steer, C.J.; Li, Z.; Chen, X.; Song, G. MicroRNA-206 prevents hepatosteatosis and hyperglycemia by facilitating insulin signaling and impairing lipogenesis. J. Hepatol. 2017, 66, 816–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Liu, X.L.; Cao, H.X.; Fan, J.G. MicroRNAs as biomarkers and regulators of nonalcoholic fatty liver disease. J. Dig. Dis. 2016, 17, 708–715. [Google Scholar] [CrossRef] [PubMed]
  54. Hsu, S.-H.; Wang, B.; Kota, J.; Yu, J.; Costinean, S.; Kutay, H.; Yu, L.; Bai, S.; La Perle, K.; Chivukula, R.R.; et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J. Clin. Investig. 2012, 122, 2871–2883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wen, J.; Friedman, J.R. miR-122 regulates hepatic lipid metabolism and tumor suppression. J. Clin. Investig. 2012, 122, 2773–2776. [Google Scholar] [CrossRef] [Green Version]
  56. Mann, J.; Chu, D.C.; Maxwell, A.; Oakley, F.; Zhu, N.; Tsukamoto, H.; Mann, D.A. MeCP2 Controls an Epigenetic Pathway That Promotes Myofibroblast Transdifferentiation and Fibrosis. Gastroenterology 2010, 138, 705–714.e4. [Google Scholar] [CrossRef] [Green Version]
  57. Roderburg, C.; Urban, G.-W.; Bettermann, K.; Vucur, M.; Zimmermann, H.W.; Schmidt, S.; Janssen, J.; Koppe, C.; Knolle, P.; Castoldi, M.; et al. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology 2010, 53, 209–218. [Google Scholar] [CrossRef] [PubMed]
  58. Sekiya, Y.; Ogawa, T.; Yoshizato, K.; Ikeda, K.; Kawada, N. Suppression of hepatic stellate cell activation by microRNA-29b. Biochem. Biophys. Res. Commun. 2011, 412, 74–79. [Google Scholar] [CrossRef]
  59. Bandyopadhyay, S.; Friedman, R.C.; Marquez, R.T.; Keck, K.; Kong, B.; Icardi, M.S.; Brown, K.E.; Burge, C.B.; Schmidt, W.N.; Wang, Y.; et al. Hepatitis C Virus Infection and Hepatic Stellate Cell Activation Downregulate miR-29: miR-29 Overexpression Reduces Hepatitis C Viral Abundance in Culture. J. Infect. Dis. 2011, 203, 1753–1762. [Google Scholar] [CrossRef]
  60. Huang, J.; Yu, X.; Fries, J.W.U.; Zhang, L.; Odenthal, M. MicroRNA Function in the Profibrogenic Interplay upon Chronic Liver Disease. Int. J. Mol. Sci. 2014, 15, 9360–9371. [Google Scholar] [CrossRef] [Green Version]
  61. Tijsen, A.J.; Van Der Made, I.; van den Hoogenhof, M.M.; Wijnen, W.J.; Van Deel, E.D.; De Groot, N.E.; Alekseev, S.; Fluiter, K.; Schroen, B.; Goumans, M.-J.; et al. The microRNA-15 family inhibits the TGFβ-pathway in the heart. Cardiovasc. Res. 2014, 104, 61–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Murakami, Y.; Toyoda, H.; Tanaka, M.; Kuroda, M.; Harada, Y.; Matsuda, F.; Tajima, A.; Kosaka, N.; Ochiya, T.; Shimotohno, K. The Progression of Liver Fibrosis Is Related with Overexpression of the miR-199 and 200 Families. PLoS ONE 2011, 6, e16081. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, X.; He, Y.; Ma, T.-T.; Huang, C.; Zhang, L.; Li, J. Participation of miR-200a in TGF-β1-mediated hepatic stellate cell activation. Mol. Cell. Biochem. 2014, 388, 11–23. [Google Scholar] [CrossRef] [PubMed]
  64. Mak, D.; Babb De Villiers, C.; Chasela, C.; Urban, M.I.; Kramvis, A. Analysis of risk factors associated with hepatocellular carcinoma in black South Africans: 2000–2012. PLoS ONE 2018, 13, e0196057. [Google Scholar] [CrossRef] [PubMed]
  65. Jiang, J.; Gusev, Y.; Aderca, I.; Mettler, T.A.; Nagorney, D.M.; Brackett, D.J.; Roberts, L.R.; Schmittgen, T.D. Association of MicroRNA Expression in Hepatocellular Carcinomas with Hepatitis Infection, Cirrhosis, and Patient Survival. Clin. Cancer Res. 2008, 14, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Furuta, M.; Kozaki, K.-I.; Tanaka, S.; Arii, S.; Imoto, I.; Inazawa, J. miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 2010, 31, 766–776. [Google Scholar] [CrossRef] [Green Version]
  67. Hatziapostolou, M.; Polytarchou, C.; Aggelidou, E.; Drakaki, A.; Poultsides, G.A.; Jaeger, S.A.; Ogata, H.; Karin, M.; Struhl, K.; Hadzopoulou-Cladaras, M.; et al. An HNF4α-miRNA Inflammatory Feedback Circuit Regulates Hepatocellular Oncogenesis. Cell 2011, 147, 1233–1247. [Google Scholar] [CrossRef] [Green Version]
  68. Murakami, Y.; Yasuda, T.; Saigo, K.; Urashima, T.; Toyoda, H.; Okanoue, T.; Shimotohno, K. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 2006, 25, 2537–2545. [Google Scholar] [CrossRef]
  69. Sartorius, K.; Makarova, J.A.; Sartorius, B.; An, P.; Winkler, C.A.; Chuturgoon, A.A.; Kramvis, A. The Regulatory Role of MicroRNA in Hepatitis-B Virus-Associated Hepatocellular Carcinoma (HBV-HCC) Pathogenesis. Cells 2019, 8, 1504. [Google Scholar] [CrossRef] [Green Version]
  70. Xie, K.-L.; Zhang, Y.-G.; Liu, J.; Zeng, Y.; Wu, H. MicroRNAs Associated With HBV Infection And HBV-related HCC. Theranostics 2014, 4, 1176–1192. [Google Scholar] [CrossRef] [Green Version]
  71. Yang, P.; Li, Q.-J.; Feng, Y.; Zhang, Y.; Markowitz, G.J.; Ning, S.; Deng, Y.; Zhao, J.; Jiang, S.; Yuan, Y.; et al. TGF-β-miR-34a-CCL22 Signaling-Induced Treg Cell Recruitment Promotes Venous Metastases of HBV-Positive Hepatocellular Carcinoma. Cancer Cell 2012, 22, 291–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Su, C.; Hou, Z.; Zhang, C.; Tian, Z.; Zhang, J. Ectopic expression of microRNA-155 enhances innate antiviral immunity against HBV infection in human hepatoma cells. Virol. J. 2011, 8, 354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, B.; Majumder, S.; Nuovo, G.; Kutay, H.; Volinia, S.; Patel, T.; Schmittgen, T.D.; Croce, C.; Ghoshal, K.; Jacob, S.T. Role of microRNA-155 at early stages of hepatocarcinogenesis induced by choline-deficient and amino acid-defined diet in C57BL/6 mice. Hepatology 2009, 50, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  74. Arataki, K.; Hayes, C.N.; Akamatsu, S.; Akiyama, R.; Abe, H.; Tsuge, M.; Miki, D.; Ochi, H.; Hiraga, N.; Aikata, H.; et al. Circulating microRNA-22 correlates with microRNA-122 and represents viral replication and liver injury in patients with chronic hepatitis B. J. Med Virol. 2013, 85, 789–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nakamura, M.; Kanda, T.; Jiang, X.; Haga, Y.; Takahashi, K.; Wu, S.; Yasui, S.; Nakamoto, S.; Yokosuka, O. Serum microRNA-122 and Wisteria floribunda agglutinin-positive Mac-2 binding protein are useful tools for liquid biopsy of the patients with hepatitis B virus and advanced liver fibrosis. PLoS ONE 2017, 12, e0177302. [Google Scholar] [CrossRef] [Green Version]
  76. Bandiera, S.; Pfeffer, S.; Baumert, T.F.; Zeisel, M.B. miR-122—A key factor and therapeutic target in liver disease. J. Hepatol. 2015, 62, 448–457. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, S.; Qiu, L.; Yan, X.; Jin, W.; Wang, Y.; Chen, L.; Wu, E.; Ye, X.; Gao, G.F.; Wang, F.; et al. Loss of microRNA 122 expression in patients with hepatitis B enhances hepatitis B virus replication through cyclin G1-modulated P53 activity. Hepatolgy 2012, 55, 730–741. [Google Scholar] [CrossRef]
  78. Qiu, L.; Fan, H.; Jin, W.; Zhao, B.; Wang, Y.; Ju, Y.; Chen, L.; Chen, Y.; Duan, Z.; Meng, S. miR-122-induced down-regulation of HO-1 negatively affects miR-122-mediated suppression of HBV. Biochem. Biophys. Res. Commun. 2010, 398, 771–777. [Google Scholar] [CrossRef]
  79. Szabo, G.; Bala, S. MicroRNAs in liver disease. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 542–552. [Google Scholar] [CrossRef] [Green Version]
  80. Fisicaro, P.; Valdatta, C.; Boni, C.; Massari, M.; Mori, C.; Zerbini, A.; Orlandini, A.; Sacchelli, L.; Missale, G.; Ferrari, C. Early kinetics of innate and adaptive immune responses during hepatitis B virus infection. Gut 2009, 58, 974–982. [Google Scholar] [CrossRef] [Green Version]
  81. Mizuguchi, Y.; Takizawa, T.; Yoshida, H.; Uchida, E. Dysregulated miRNA in progression of hepatocellular carcinoma: A systematic review. Hepatol. Res. 2016, 46, 391–406. [Google Scholar] [CrossRef] [PubMed]
  82. Bertoletti, A.; Kennedy, P.T.F. The immune tolerant phase of chronic HBV infection: New perspectives on an old concept. Cell. Mol. Immunol. 2014, 12, 258–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Bertoletti, A.; Ferrari, C. Adaptive immunity in HBV infection. J. Hepatol. 2016, 64, S71–S83. [Google Scholar] [CrossRef] [PubMed]
  84. Busca, A.; Kumar, A. Innate immune responses in hepatitis B virus (HBV) infection. Virol. J. 2014, 11, 22. [Google Scholar] [CrossRef] [Green Version]
  85. Torres, H.A.; Shigle, T.L.; Hammoudi, N.; Link, J.T.; Samaniego, F.; Kaseb, A.; Mallet, V. The oncologic burden of hepatitis C virus infection: A clinical perspective. CA A Cancer J. Clin. 2017, 67, 411–431. [Google Scholar] [CrossRef]
  86. Bachofner, J.; Valli, P.V.; Bergamin, I.; Kröger, A.; Künzler, P.; Baserga, A.; Braun, D.L.; Seifert, B.; Moncsek, A.; Fehr, J.; et al. Excellent outcome of direct antiviral treatment for chronic hepatitis C in Switzerland. Swiss Med Wkly. 2018, 148, w14560. [Google Scholar] [CrossRef] [Green Version]
  87. Shoukry, N.H. Hepatitis C Vaccines, Antibodies, and T Cells. Front. Immunol. 2018, 9, 1480. [Google Scholar] [CrossRef]
  88. Ura, S.; Honda, M.; Yamashita, T.; Ueda, T.; Takatori, H.; Nishino, R.; Sunakozaka, H.; Sakai, Y.; Horimoto, K.; Kaneko, S. Differential microRNA expression between hepatitis B and hepatitis C leading disease progression to hepatocellular carcinoma. Hepatolgy 2009, 49, 1098–1112. [Google Scholar] [CrossRef]
  89. Steuerwald, N.M.; Parsons, J.C.; Bennett, K.; Bates, T.C.; Bonkovsky, H.L. Parallel microRNA and mRNA expression profiling of (genotype 1b) human hepatoma cells expressing hepatitis C virus. Liver Int. 2010, 30, 1490–1504. [Google Scholar] [CrossRef]
  90. Kerr, T.A.; Korenblat, K.M.; Davidson, N.O. MicroRNAs and liver disease. Transl. Res. 2011, 157, 241–252. [Google Scholar] [CrossRef] [Green Version]
  91. Pedersen, I.M.; Cheng, G.; Wieland, S.; Volinia, S.; Croce, C.M.; Chisari, F.V.; David, M. Interferon modulation of cellular microRNAs as an antiviral mechanism. Nature 2007, 449, 919–922. [Google Scholar] [CrossRef]
  92. Peng, X.; Li, Y.; Walters, K.-A.; Rosenzweig, E.R.; Lederer, S.L.; Aicher, L.D.; Proll, S.; Katze, M.G. Computational identification of hepatitis C virus associated microRNA-mRNA regulatory modules in human livers. BMC Genom. 2009, 10, 373. [Google Scholar] [CrossRef] [Green Version]
  93. Lanford, R.E.; Hildebrandt-Eriksen, E.S.; Petri, A.; Persson, R.; Lindow, M.; Munk, M.E.; Kauppinen, S.; Orum, H. Therapeutic Silencing of MicroRNA-122 in Primates with Chronic Hepatitis C Virus Infection. Science 2009, 327, 198–201. [Google Scholar] [CrossRef] [Green Version]
  94. Jopling, C.L.; Yi, M.; Lancaster, A.M.; Lemon, S.M.; Sarnow, P. Modulation of Hepatitis C Virus RNA Abundance by a Liver-Specific MicroRNA. Science 2005, 309, 1577–1581. [Google Scholar] [CrossRef] [Green Version]
  95. Banaudha, K.; Kaliszewski, M.; Korolnek, T.; Florea, L.; Yeung, M.L.; Jeang, K.-T.; Kumar, A. MicroRNA silencing of tumor suppressor DLC-1 promotes efficient hepatitis C virus replication in primary human hepatocytes. Hepatolgy 2011, 53, 53–61. [Google Scholar] [CrossRef]
  96. Ishida, H.; Tatsumi, T.; Hosui, A.; Nawa, T.; Kodama, T.; Shimizu, S.; Hikita, H.; Hiramatsu, N.; Kanto, T.; Hayashi, N.; et al. Alterations in microRNA expression profile in HCV-infected hepatoma cells: Involvement of miR-491 in regulation of HCV replication via the PI3 kinase/Akt pathway. Biochem. Biophys. Res. Commun. 2011, 412, 92–97. [Google Scholar] [CrossRef]
  97. Murakami, Y.; Aly, H.H.; Tajima, A.; Inoue, I.; Shimotohno, K. Regulation of the hepatitis C virus genome replication by miR-199a. J. Hepatol. 2009, 50, 453–460. [Google Scholar] [CrossRef]
  98. Hou, W.; Tian, Q.; Zheng, J.; Bonkovsky, H.L. MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expression in human hepatoma cells expressing hepatitis C viral proteins. Hepatolgy 2010, 51, 1494–1504. [Google Scholar] [CrossRef] [Green Version]
  99. Cheng, J.-C.; Yeh, Y.-J.; Tseng, C.-P.; Hsu, S.-D.; Chang, Y.-L.; Sakamoto, N.; Huang, H.-D. Let-7b is a novel regulator of hepatitis C virus replication. Cell. Mol. Life Sci. 2012, 69, 2621–2633. [Google Scholar] [CrossRef]
  100. Chowdhury, J.B.; Shrivastava, S.; Steele, R.; Di Bisceglie, A.M.; Ray, R.; Ray, R.B. Hepatitis C Virus Infection Modulates Expression of Interferon Stimulatory Gene IFITM1 by Upregulating miR-130A. J. Virol. 2012, 86, 10221–10225. [Google Scholar] [CrossRef] [Green Version]
  101. Shirasaki, T.; Honda, M.; Shimakami, T.; Horii, R.; Yamashita, T.; Sakai, Y.; Sakai, A.; Okada, H.; Watanabe, R.; Murakami, S.; et al. MicroRNA-27a Regulates Lipid Metabolism and Inhibits Hepatitis C Virus Replication in Human Hepatoma Cells. J. Virol. 2013, 87, 5270–5286. [Google Scholar] [CrossRef] [Green Version]
  102. Li, H.-D.; Du, X.-S.; Huang, H.-M.; Chen, X.; Yang, Y.; Huang, C.; Meng, X.-M.; Li, J. Noncoding RNAs in alcoholic liver disease. J. Cell. Physiol. 2019, 234, 14709–14720. [Google Scholar] [CrossRef]
  103. Mandrekar, P.; Szabo, G. Signalling pathways in alcohol-induced liver inflammation. J. Hepatol. 2009, 50, 1258–1266. [Google Scholar] [CrossRef] [Green Version]
  104. Gao, B.; Bataller, R. Alcoholic Liver Disease: Pathogenesis and New Therapeutic Targets. Gastroenterology 2011, 141, 1572–1585. [Google Scholar] [CrossRef] [Green Version]
  105. Tang, Y.; Banan, A.; Forsyth, C.B.; Fields, J.Z.; Lau, C.K.; Zhang, L.J.; Keshavarzian, A. Effect of Alcohol on miR-212 Expression in Intestinal Epithelial Cells and Its Potential Role in Alcoholic Liver Disease. Alcohol. Clin. Exp. Res. 2008, 32, 355–364. [Google Scholar] [CrossRef]
  106. Yeligar, S.; Tsukamoto, H.; Kalra, V.K. Ethanol-Induced Expression of ET-1 and ET-BR in Liver Sinusoidal Endothelial Cells and Human Endothelial Cells Involves Hypoxia-Inducible Factor-1α and MicroRNA-199. J. Immunol. 2009, 183, 5232–5243. [Google Scholar] [CrossRef] [Green Version]
  107. Li, M.; He, Y.; Zhou, Z.; Ramirez, T.; Gao, Y.; Gao, Y.; Ross, R.A.; Cao, H.; Cai, Y.; Xu, M.; et al. MicroRNA-223 ameliorates alcoholic liver injury by inhibiting the IL-6–p47phox–oxidative stress pathway in neutrophils. Gut 2017, 66, 705–715. [Google Scholar] [CrossRef] [Green Version]
  108. Yin, H.; Liang, X.; Jogasuria, A.; Davidson, N.O.; You, M. miR-217 Regulates Ethanol-Induced Hepatic Inflammation by Disrupting Sirtuin 1–Lipin-1 Signaling. Am. J. Pathol. 2015, 185, 1286–1296. [Google Scholar] [CrossRef] [Green Version]
  109. Wan, Y.; McDaniel, K.; Wu, N.; Ramos-Lorenzo, S.; Glaser, T.; Venter, J.; Francis, H.; Kennedy, L.; Sato, K.; Zhou, T.; et al. Regulation of Cellular Senescence by miR-34a in Alcoholic Liver Injury. Am. J. Pathol. 2017, 187, 2788–2798. [Google Scholar] [CrossRef] [Green Version]
  110. Esau, C.; Davis, S.; Murray, S.F.; Yu, X.X.; Pandey, S.K.; Pear, M.; Watts, L.; Booten, S.L.; Graham, M.; McKay, R.; et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006, 3, 87–98. [Google Scholar] [CrossRef] [Green Version]
  111. McDaniel, K.; Huang, L.; Sato, K.; Wu, N.; Annable, T.; Zhou, T.; Ramos-Lorenzo, S.; Wan, Y.; Huang, Q.; Francis, H.; et al. The let-7/Lin28 axis regulates activation of hepatic stellate cells in alcoholic liver injury. J. Biol. Chem. 2017, 292, 11336–11347. [Google Scholar] [CrossRef] [Green Version]
  112. Nuño-Lámbarri, N.; Domínguez-Pérez, M.; Baulies-Domenech, A.; Monte, M.J.; Marin, J.J.G.; Rosales-Cruz, P.; Souza, V.; Miranda, R.U.; Bucio, L.; Montalvo-Jave, E.E.; et al. Liver Cholesterol Overload Aggravates Obstructive Cholestasis by Inducing Oxidative Stress and Premature Death in Mice. Oxidative Med. Cell. Longev. 2016, 2016, 9895176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Bernal-Reyes, R.; Castro-Narro, G.; Malé-Velázquez, R.; Carmona-Sánchez, R.; González-Huezo, M.; García-Juárez, I.; Chávez-Tapia, N.; Aguilar-Salinas, C.; Aiza-Haddad, I.; Ballesteros-Amozurrutia, M.; et al. Consenso mexicano de la enfermedad por hígado graso no alcohólico. Rev. Gastroenterol. México 2019, 84, 69–99. [Google Scholar] [CrossRef] [PubMed]
  114. Pirola, C.J.; Fernandez Gianotti, T.; Castano, G.O.; Mallardi, P.; San Martino, J.; Mora Gonzalez Lopez Ledesma, M.; Flichman, D.M.; Mirshahi, F.; Sanyal, A.J.; Sookoian, S.C. Circulating microRNA signature in non-alcoholic fatty liver disease: From serum non-coding RNAs to liver histology and disease pathogenesis. Gut 2015, 64, 8001–8002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wu, F.-L.; Jin, W.-B.; Li, J.-H.; Guo, A.-G. Targets for human encoded microRNAs in HBV genes. Virus Genes 2011, 42, 157–161. [Google Scholar] [CrossRef]
  116. Coll, M.; El Taghdouini, A.; Perea, L.; Mannaerts, I.; Vila-Casadesús, M.; Blaya, D.; Rodrigo-Torres, D.; Affò, S.; Morales-Ibanez, O.; Graupera, I.; et al. Integrative miRNA and Gene Expression Profiling Analysis of Human Quiescent Hepatic Stellate Cells. Sci. Rep. 2015, 5, 11549. [Google Scholar] [CrossRef] [Green Version]
  117. Tomita, K.; Teratani, T.; Suzuki, T.; Shimizu, M.; Sato, H.; Narimatsu, K.; Okada, Y.; Kurihara, C.; Irie, R.; Yokoyama, H.; et al. Free cholesterol accumulation in hepatic stellate cells: Mechanism of liver fibrosis aggravation in nonalcoholic steatohepatitis in mice. Hepatology 2014, 59, 154–169. [Google Scholar] [CrossRef]
  118. Vinciguerra, M.; Sgroi, A.; Veyrat-Durebex, C.; Rubbia-Brandt, L.; Buhler, L.H.; Foti, M. Unsaturated fatty acids inhibit the expression of tumor suppressor phosphatase and tensin homolog (PTEN) via microRNA-21 up-regulation in hepatocytes. Hepatology 2009, 49, 1176–1184. [Google Scholar] [CrossRef]
  119. O’Connell, R.M.; Rao, D.S.; Chaudhuri, A.A.; Boldin, M.P.; Taganov, K.D.; Nicoll, J.; Paquette, R.L.; Baltimore, D. Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder. J. Exp. Med. 2008, 205, 585–594. [Google Scholar] [CrossRef]
  120. Feng, X.; Jiang, J.; Shi, S.; Xie, H.; Zhou, L.; Zheng, S. Knockdown of miR-25 increases the sensitivity of liver cancer stem cells to TRAIL-induced apoptosis via PTEN/PI3K/Akt/Bad signaling pathway. Int. J. Oncol. 2016, 49, 2600–2610. [Google Scholar] [CrossRef] [Green Version]
  121. Iwai, N.; Yasui, K.; Tomie, A.; Gen, Y.; Terasaki, K.; Kitaichi, T.; Soda, T.; Yamada, N.; Dohi, O.; Seko, Y.; et al. Oncogenic miR-96-5p inhibits apoptosis by targeting the caspase-9 gene in hepatocellular carcinoma. Int. J. Oncol. 2018, 53, 237–245. [Google Scholar] [CrossRef] [Green Version]
  122. Koenig, A.; Barajas, J.M.; Guerrero, M.J.; Ghoshal, K. A Comprehensive Analysis of Argonaute-CLIP Data Identifies Novel, Conserved and Species-Specific Targets of miR-21 in Human Liver and Hepatocellular Carcinoma. Int. J. Mol. Sci. 2018, 19, 851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Su, S.-G.; Yang, M.; Zhang, M.-F.; Peng, Q.-Z.; Li, M.-Y.; Liu, L.; Bao, S. miR-107-mediated decrease of HMGCS2 indicates poor outcomes and promotes cell migration in hepatocellular carcinoma. Int. J. Biochem. Cell Biol. 2017, 91, 53–59. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, Y.; Chen, F.; Zhao, M.; Yang, Z.; Zhang, S.; Ye, L.; Gao, H.; Zhang, X. MiR-107 suppresses proliferation of hepatoma cells through targeting HMGA2 mRNA 3′UTR. Biochem. Biophys. Res. Commun. 2016, 480, 455–460. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, W.; Dou, C.; Wang, Y.; Jia, Y.; Li, C.; Zheng, X.; Tu, K. MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7. Oncol. Rep. 2015, 34, 2576–2584. [Google Scholar] [CrossRef] [Green Version]
  126. Khan, S.; Ayub, H.; Khan, T.; Wahid, F. MicroRNA biogenesis, gene silencing mechanisms and role in breast, ovarian and prostate cancer. Biochimie 2019, 167, 12–24. [Google Scholar] [CrossRef]
  127. Morishita, A.; Iwama, H.; Fujihara, S.; Sakamoto, T.; Fujita, K.; Tani, J.; Miyoshi, H.; Yoneyama, H.; Himoto, T.; Masaki, T. MicroRNA profiles in various hepatocellular carcinoma cell lines. Oncol. Lett. 2016, 12, 1687–1692. [Google Scholar] [CrossRef] [Green Version]
  128. Wang, Y.; Lu, Y.; Toh, S.T.; Sung, W.-K.; Tan, P.; Chow, P.; Chung, A.Y.; Jooi, L.L.; Lee, C.G.L. Lethal-7 is down-regulated by the hepatitis B virus x protein and targets signal transducer and activator of transcription 3. J. Hepatol. 2010, 53, 57–66. [Google Scholar] [CrossRef]
  129. Wang, Z.; Lin, S.; Li, J.J.; Xu, Z.; Yao, H.; Zhu, X.; Xie, D.; Shen, Z.; Sze, J.; Li, K.; et al. MYC Protein Inhibits Transcription of the MicroRNA ClusterMC-let-7a-1∼let-7d via Noncanonical E-box. J. Biol. Chem. 2011, 286, 39703–39714. [Google Scholar] [CrossRef] [Green Version]
  130. Tsang, W.P.; Kwok, T.T. Let-7a microRNA suppresses therapeutics-induced cancer cell death by targeting caspase-3. Apoptosis 2008, 13, 1215–1222. [Google Scholar] [CrossRef]
  131. Di Fazio, P.; Montalbano, R.; Neureiter, D.; Alinger, B.; Schmidt, A.; Merkel, A.L.; Quint, K.; Ocker, M. Downregulation of HMGA2 by the pan-deacetylase inhibitor panobinostat is dependent on hsa-let-7b expression in liver cancer cell lines. Exp. Cell Res. 2012, 318, 1832–1843. [Google Scholar] [CrossRef]
  132. Au, S.L.-K.; Wong, C.C.-L.; Lee, J.M.-F.; Fan, D.N.-Y.; Tsang, F.H.; Ng, I.O.-L.; Wong, C.-M. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatology 2012, 56, 622–631. [Google Scholar] [CrossRef]
  133. Zhu, X.-M.; Wu, L.-J.; Xu, J.; Yang, R.; Wu, F.-S. Let-7c MicroRNA Expression and Clinical Significance in Hepatocellular Carcinoma. J. Int. Med Res. 2011, 39, 2323–2329. [Google Scholar] [CrossRef] [Green Version]
  134. Shah, Y.M.; Morimura, K.; Yang, Q.; Tanabe, T.; Takagi, M.; Gonzalez, F.J. Peroxisome Proliferator-Activated Receptor α Regulates a MicroRNA-Mediated Signaling Cascade Responsible for Hepatocellular Proliferation. Mol. Cell. Biol. 2007, 27, 4238–4247. [Google Scholar] [CrossRef] [Green Version]
  135. Shimizu, S.; Takehara, T.; Hikita, H.; Kodama, T.; Miyagi, T.; Hosui, A.; Tatsumi, T.; Ishida, H.; Noda, T.; Nagano, H.; et al. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. J. Hepatol. 2010, 52, 698–704. [Google Scholar] [CrossRef]
  136. Ji, J.; Zhao, L.; Budhu, A.; Forgues, M.; Jia, H.-L.; Qin, L.-X.; Ye, Q.-H.; Yu, J.; Shi, X.; Tang, Z.-Y.; et al. Let-7g targets collagen type I α2 and inhibits cell migration in hepatocellular carcinoma. J. Hepatol. 2010, 52, 690–697. [Google Scholar] [CrossRef] [Green Version]
  137. Lan, F.-F.; Wang, H.; Chen, Y.-C.; Chan, C.-Y.; Ng, S.S.; Li, K.; Xie, D.; He, M.-L.; Lin, M.C.; Kung, H.-F. Hsa-let-7g inhibits proliferation of hepatocellular carcinoma cells by downregulation of c-Myc and upregulation of p16INK4A. Int. J. Cancer 2011, 128, 319–331. [Google Scholar] [CrossRef]
  138. Li, N.; Yang, P.; Li, H.; Cheng, P.; Zhang, L.; Wei, D.; Su, X.; Peng, J.; Gao, H.; Tan, Y.; et al. MicroRNA-1 inhibits proliferation of hepatocarcinoma cells by targeting endothelin-1. Life Sci. 2012, 91, 440–447. [Google Scholar] [CrossRef]
  139. Fang, Y.; Xue, J.-L.; Shen, Q.; Chen, J.; Tian, L. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology 2012, 55, 1852–1862. [Google Scholar] [CrossRef]
  140. Yan, Y.; Luo, Y.-C.; Wan, H.-Y.; Wang, J.; Zhang, P.-P.; Liu, M.; Li, X.; Li, S.; Tang, H. MicroRNA-10a is involved in the metastatic process by regulating Eph tyrosine kinase receptor A4-Mediated epithelial-mesenchymal transition and adhesion in hepatoma cells. Hepatology 2013, 57, 667–677. [Google Scholar] [CrossRef] [Green Version]
  141. Maurel, M.; Jalvy, S.; Ladeiro, Y.; Combe, C.; Vachet, L.; Sagliocco, F.; Bioulac-Sage, P.; Pitard, V.; Jacquemin-Sablon, H.; Zucman-Rossi, J.; et al. A functional screening identifies five micrornas controlling glypican-3: Role of mir-1271 down-regulation in hepatocellular carcinoma. Hepatology 2013, 57, 195–204. [Google Scholar] [CrossRef]
  142. Wang, Y.; Jiang, L.; Ji, X.; Yang, B.; Zhang, Y.; Fu, X.-D. Hepatitis B Viral RNA Directly Mediates Down-regulation of the Tumor Suppressor MicroRNA miR-15a/miR-16-1 in Hepatocytes. J. Biol. Chem. 2013, 288, 18484–18493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zhang, Y.; Huang, F.; Wang, J.; Peng, L.; Luo, H. MiR-15b mediates liver cancer cells proliferation through targeting BCL-2. Int. J. Clin. Exp. Pathol. 2015, 8, 15677–15683. [Google Scholar] [PubMed]
  144. Ladeiro, Y.; Couchy, G.; Balabaud, C.; Bioulac-Sage, P.; Pelletier, L.; Rebouissou, S.; Zucman-Rossi, J. MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology 2008, 47, 1955–1963. [Google Scholar] [CrossRef]
  145. Yang, X.; Guo, H.-M.; Zhang, X.-F.; Jia, H.-L.; Qin, Y.; Zhu, X.-C.; Gao, X.-M.; Qiao, P.; Zheng, Y.; Sheng, Y.; et al. MicroRNA-26a suppresses tumor growth and metastasis of human hepatocellular carcinoma by targeting interleukin-6-Stat3 pathway. Hepatology 2013, 58, 158–170. [Google Scholar] [CrossRef] [PubMed]
  146. Kota, J.; Chivukula, R.R.; O’Donnell, K.A.; Wentzel, E.A.; Montgomery, C.L.; Hwang, H.-W.; Chang, T.-C.; Vivekanandan, P.; Torbenson, M.; Clark, K.R.; et al. Therapeutic microRNA Delivery Suppresses Tumorigenesis in a Murine Liver Cancer Model. Cell 2009, 137, 1005–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Zhu, Y.; Lu, Y.; Zhang, Q.; Liu, J.-J.; Li, T.-J.; Yang, J.-R.; Zeng, C.; Zhuang, S.-M. MicroRNA-26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb protein. Nucleic Acids Res. 2011, 40, 4615–4625. [Google Scholar] [CrossRef] [PubMed]
  148. Zhu, X.-C.; Dong, Q.; Zhang, X.-F.; Deng, B.; Jia, H.; Ye, Q.; Qin, L.-X.; Wu, X.Z. microRNA-29a suppresses cell proliferation by targeting SPARC in hepatocellular carcinoma. Int. J. Mol. Med. 2012, 30, 1321–1326. [Google Scholar] [CrossRef]
  149. Fang, J.-H.; Zhou, H.-C.; Zeng, C.; Yang, J.; Liu, Y.; Huang, X.; Zhang, J.-P.; Guan, X.-Y.; Zhuang, S.-M. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology 2011, 54, 1729–1740. [Google Scholar] [CrossRef]
  150. Bae, H.J.; Noh, J.H.; Kim, J.K.; Eun, J.W.; Jung, K.H.; Kim, M.G.; Chang, Y.G.; Shen, Q.; Kim, S.-J.; Park, W.S.; et al. MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene 2014, 33, 2557–2567. [Google Scholar] [CrossRef] [Green Version]
  151. Zhao, G.; Han, C.; Zhang, Z.; Wang, L.; Xu, J. Increased expression of microRNA-31-5p inhibits cell proliferation, migration, and invasion via regulating Sp1 transcription factor in HepG2 hepatocellular carcinoma cell line. Biochem. Biophys. Res. Commun. 2017, 490, 371–377. [Google Scholar] [CrossRef] [PubMed]
  152. Tian, Q.; Xiao, Y.; Wu, Y.; Liu, Y.; Song, Z.; Gao, W.; Zhang, J.; Yang, J.; Zhang, Y.; Guo, T.; et al. MicroRNA-33b suppresses the proliferation and metastasis of hepatocellular carcinoma cells through the inhibition of Sal-like protein 4 expression. Int. J. Mol. Med. 2016, 38, 1587–1595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Li, N.; Fu, H.; Tie, Y.; Hu, Z.; Kong, W.; Wu, Y.; Zheng, X. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett. 2009, 275, 44–53. [Google Scholar] [CrossRef] [PubMed]
  154. Zhang, J.-J.; Chen, J.-T.; Hua, L.; Yao, K.-H.; Wang, C.-Y. miR-98 inhibits hepatocellular carcinoma cell proliferation via targeting EZH2 and suppressing Wnt/β-catenin signaling pathway. Biomed. Pharmacother. 2017, 85, 472–478. [Google Scholar] [CrossRef] [PubMed]
  155. Petrelli, A.; Perra, A.; Schernhuber, K.; Cargnelutti, M.; Salvi, A.; Migliore, C.; Ghiso, E.; Benetti, A.; Barlati, S.; Ledda-Columbano, G.M.; et al. Sequential analysis of multistage hepatocarcinogenesis reveals that miR-100 and PLK1 dysregulation is an early event maintained along tumor progression. Oncogene 2012, 31, 4517–4526. [Google Scholar] [CrossRef] [Green Version]
  156. Li, D.; Liu, X.; Lin, L.; Hou, J.; Li, N.; Wang, C.; Wang, P.; Zhang, Q.; Zhang, P.; Zhou, W.; et al. MicroRNA-99a Inhibits Hepatocellular Carcinoma Growth and Correlates with Prognosis of Patients with Hepatocellular Carcinoma. J. Biol. Chem. 2011, 286, 36677–36685. [Google Scholar] [CrossRef] [Green Version]
  157. Wang, L.; Zhang, X.; Jia, L.; Hu, S.; Zhao, J.; Yang, J.-D.; Wen, W.-H.; Wang, Z.; Wang, T.; Zhao, J.; et al. c-Myc-mediated epigenetic silencing of MicroRNA-101 contributes to dysregulation of multiple pathways in hepatocellular carcinoma. Hepatology 2014, 59, 1850–1863. [Google Scholar] [CrossRef]
  158. Su, H.; Yang, J.-R.; Xu, T.; Huang, J.; Xu, L.; Yuan, Y.; Zhuang, S.-M. MicroRNA-101, Down-regulated in Hepatocellular Carcinoma, Promotes Apoptosis and Suppresses Tumorigenicity. Cancer Res. 2009, 69, 1135–1142. [Google Scholar] [CrossRef] [Green Version]
  159. Li, S.; Fu, H.; Wang, Y.; Tie, Y.; Xing, R.; Zhu, J.; Sun, Z.; Wei, L.; Zheng, X. MicroRNA-101 regulates expression of the v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) oncogene in human hepatocellular carcinoma. Hepatology 2009, 49, 1194–1202. [Google Scholar] [CrossRef]
  160. Lin, C.J.-F.; Gong, H.-Y.; Tseng, H.-C.; Wang, W.-L.; Wu, J.-L. miR-122 targets an anti-apoptotic gene, Bcl-w, in human hepatocellular carcinoma cell lines. Biochem. Biophys. Res. Commun. 2008, 375, 315–320. [Google Scholar] [CrossRef]
  161. Kong, G.; Zhang, J.; Zhang, S.; Shan, C.; Ye, L.; Zhang, X. Upregulated MicroRNA-29a by Hepatitis B Virus X Protein Enhances Hepatoma Cell Migration by Targeting PTEN in Cell Culture Model. PLoS ONE 2011, 6, e19518. [Google Scholar] [CrossRef] [PubMed]
  162. Reddi, H.V.; Madde, P.; Milosevic, D.; Hackbarth, J.S.; Algeciras-Schimnich, A.; McIver, B.; Grebe, S.K.; Eberhardt, N.L. The Putative PAX8/PPARγγ Fusion Oncoprotein Exhibits Partial Tumor Suppressor Activity through Up-Regulation of Micro-RNA-122 and Dominant-Negative PPAR Activity. Genes Cancer 2011, 2, 46–55. [Google Scholar] [CrossRef] [PubMed]
  163. Xu, J.; Zhu, X.; Wu, L.; Yang, R.; Yang, Z.; Wang, Q.; Wu, F. MicroRNA-122 suppresses cell proliferation and induces cell apoptosis in hepatocellular carcinoma by directly targeting Wnt/β-catenin pathway. Liver Int. 2012, 32, 752–760. [Google Scholar] [CrossRef] [PubMed]
  164. Lang, Q.; Ling, C. MiR-124 suppresses cell proliferation in hepatocellular carcinoma by targeting PIK3CA. Biochem. Biophys. Res. Commun. 2012, 426, 247–252. [Google Scholar] [CrossRef] [PubMed]
  165. Bi, Q.; Tang, S.; Xia, L.; Du, R.; Fan, R.; Gao, L.; Jin, J.; Liang, S.; Chen, Z.; Xu, G.; et al. Ectopic Expression of MiR-125a Inhibits the Proliferation and Metastasis of Hepatocellular Carcinoma by Targeting MMP11 and VEGF. PLoS ONE 2012, 7, e40169. [Google Scholar] [CrossRef]
  166. Kim, J.K.; Noh, J.H.; Jung, K.H.; Eun, J.W.; Bae, H.J.; Kim, M.G.; Chang, Y.G.; Shen, Q.; Park, W.S.; Lee, J.Y.; et al. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology 2013, 57, 1055–1067. [Google Scholar] [CrossRef]
  167. Zhao, A.; Zeng, Q.; Xie, X.; Zhou, J.; Yue, W.; Li, Y.; Pei, X. MicroRNA-125b Induces Cancer Cell Apoptosis Through Suppression of Bcl-2 Expression. J. Genet. Genom. 2012, 39, 29–35. [Google Scholar] [CrossRef]
  168. Alpini, G.; Glaser, S.S.; Zhang, J.-P.; Francis, H.; Han, Y.; Gong, J.; Stokes, A.; Francis, T.; Hughart, N.; Hubble, L.; et al. Regulation of placenta growth factor by microRNA-125b in hepatocellular cancer. J. Hepatol. 2011, 55, 1339–1345. [Google Scholar] [CrossRef] [Green Version]
  169. Liang, L.; Wong, C.-M.; Ying, Q.; Fan, D.N.-Y.; Huang, S.; Ding, J.; Yao, J.; Yan, M.; Li, J.; Yao, M.; et al. MicroRNA-125b suppressesed human liver cancer cell proliferation and metastasis by directly targeting oncogene LIN28B2. Hepatology 2010, 52, 1731–1740. [Google Scholar] [CrossRef]
  170. Fan, D.N.-Y.; Tsang, F.H.-C.; Tam, A.H.-K.; Au, S.L.-K.; Wong, C.C.-L.; Wei, L.; Lee, J.M.-F.; He, X.; Ng, I.O.-L.; Wong, C.-M. Histone lysine methyltransferase, suppressor of variegation 3-9 homolog 1, promotes hepatocellular carcinoma progression and is negatively regulated by microRNA-125b. Hepatology 2013, 57, 637–647. [Google Scholar] [CrossRef] [Green Version]
  171. Jing, B.-Q.; Ou, Y.; Zhao, L.; Xie, Q.; Zhang, Y.-X. Experimental study on the prevention of liver cancer angiogenesis via miR-126. Eur. Rev. Med Pharmacol. Sci. 2017, 21, 5096–5100. [Google Scholar] [CrossRef] [PubMed]
  172. Cui, S.; Sun, Y.; Liu, Y.; Liu, C.; Wang, J.; Hao, G.; Sun, Q. MicroRNA-137 has a suppressive role in liver cancer via targeting EZH2. Mol. Med. Rep. 2017, 16, 9494–9502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Wong, Q.W.-L.; Ching, A.K.-K.; Chan, A.W.-H.; Choy, K.-W.; To, K.-F.; Lai, P.B.-S.; Wong, N. MiR-222 Overexpression Confers Cell Migratory Advantages in Hepatocellular Carcinoma through Enhancing AKT Signaling. Clin. Cancer Res. 2010, 16, 867–875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Fan, Q.; He, M.; Deng, X.; Wu, W.K.; Zhao, L.; Tang, J.; Wen, G.; Sun, X.; Liu, Y. Derepression of c-Fos caused by MicroRNA-139 down-regulation contributes to the metastasis of human hepatocellular carcinoma. Cell Biochem. Funct. 2013, 31, 319–324. [Google Scholar] [CrossRef] [PubMed]
  175. Yang, H.; Fang, F.; Chang, R.; Yang, L.-Y. MicroRNA-140-5p suppresses tumor growth and metastasis by targeting transforming growth factor β receptor 1 and fibroblast growth factor 9 in hepatocellular carcinoma. Hepatology 2013, 58, 205–217. [Google Scholar] [CrossRef] [PubMed]
  176. Takata, A.; Otsuka, M.; Yoshikawa, T.; Kishikawa, T.; Hikiba, Y.; Obi, S.; Goto, T.; Kang, Y.J.; Maeda, S.; Yoshida, H.; et al. MicroRNA-140 acts as a liver tumor suppressor by controlling NF-κB activity by directly targeting DNA methyltransferase 1 (Dnmt1) expression. Hepatology 2012, 57, 162–170. [Google Scholar] [CrossRef]
  177. Su, F.; Zhao, J.; Qin, S.; Wang, R.; Li, Y.; Wang, Q.; Tan, Y.; Jin, H.; Zhu, F.; Ou, Y.; et al. Over-expression of Thrombospondin 4 correlates with loss of miR-142 and contributes to migration and vascular invasion of advanced hepatocellular carcinoma. Oncotarget 2017, 8, 23277–23288. [Google Scholar] [CrossRef] [Green Version]
  178. Yu, Q.; Xiang, L.; Yin, L.; Liu, X.; Yang, D.; Zhou, J. Loss-of-function of miR-142 by hypermethylation promotes TGF-β-mediated tumour growth and metastasis in hepatocellular carcinoma. Cell Prolif. 2017, 50, e12384. [Google Scholar] [CrossRef] [Green Version]
  179. Hua, S.; Liu, C.; Liu, L.; Wu, D. miR-142-3p inhibits aerobic glycolysis and cell proliferation in hepatocellular carcinoma via targeting LDHA. Biochem. Biophys. Res. Commun. 2018, 496, 947–954. [Google Scholar] [CrossRef]
  180. Bao, H.; Li, X.; Li, H.; Xing, H.; Xu, B.; Zhang, X.; Liu, Z. MicroRNA-144 inhibits hepatocellular carcinoma cell proliferation, invasion and migration by targeting ZFX. J. Biosci. 2017, 42, 103–111. [Google Scholar] [CrossRef]
  181. Law, P.T.-Y.; Ching, A.K.-K.; Chan, A.W.H.; Wong, Q.W.-L.; Wong, C.-K.; To, K.-F.; Wong, N. MiR-145 modulates multiple components of the insulin-like growth factor pathway in hepatocellular carcinoma. Carcinogenesis 2012, 33, 1134–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Zhang, W.; Jia, Y.; Liu, H.; Zhuang, Q.; Xu, S.; Yang, Z.; Li, J.; Lou, J. Tumorigenicity of cancer stem-like cells derived from hepatocarcinoma is regulated by microRNA-145. Oncol. Rep. 2012, 27, 1865–1872. [Google Scholar] [CrossRef] [PubMed]
  183. Gailhouste, L.; Gomez-Santos, L.; Hagiwara, K.; Hatada, I.; Kitagawa, N.; Kawaharada, K.; Thirion, M.; Kosaka, N.; Takahashi, R.-U.; Shibata, T.; et al. miR-148a plays a pivotal role in the liver by promoting the hepatospecific phenotype and suppressing the invasiveness of transformed cells. Hepatology 2013, 58, 1153–1165. [Google Scholar] [CrossRef]
  184. Xu, X.; Fan, Z.; Kang, L.; Han, J.; Jiang, C.; Zheng, X.; Zhu, Z.; Jiao, H.; Lin, J.; Jiang, K.; et al. Hepatitis B virus X protein represses miRNA-148a to enhance tumorigenesis. J. Clin. Investig. 2013, 123, 630–645. [Google Scholar] [CrossRef] [Green Version]
  185. Zhang, J.-P.; Zeng, C.; Xu, L.; Gong, J.; Fang, J.-H.; Zhuang, S.-M. MicroRNA-148a suppresses the epithelial–mesenchymal transition and metastasis of hepatoma cells by targeting Met/Snail signaling. Oncogene 2013, 33, 4069–4076. [Google Scholar] [CrossRef]
  186. Han, H.; Sun, D.; Li, W.; Shen, H.; Zhu, Y.; Li, C.; Chen, Y.; Lu, L.; Li, W.; Zhang, J.; et al. A c-Myc-MicroRNA functional feedback loop affects hepatocarcinogenesis. Hepatology 2013, 57, 2378–2389. [Google Scholar] [CrossRef]
  187. Huang, J.; Wang, Y.; Guo, Y.; Sun, S. Down-regulated microRNA-152 induces aberrant DNA methylation in hepatitis B virus-related hepatocellular carcinoma by targeting DNA methyltransferase 1. Hepatology 2010, 52, 60–70. [Google Scholar] [CrossRef] [PubMed]
  188. Bala, S.; Szabo, G. MicroRNA Signature in Alcoholic Liver Disease. Int. J. Hepatol. 2012, 2012, 498232. [Google Scholar] [CrossRef] [Green Version]
  189. Xu, T.; Zhu, Y.; Xiong, Y.; Ge, Y.-Y.; Yun, J.-P.; Zhuang, S.-M. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology 2009, 50, 113–121. [Google Scholar] [CrossRef]
  190. Yang, X.; Yu, J.; Yin, J.; Xiang, Q.; Tang, H.; Lei, X. MiR-195 regulates cell apoptosis of human hepatocellular carcinoma cells by targeting LATS2. Die Pharm. 2012, 67, 645–651. [Google Scholar]
  191. Fornari, F.; Milazzo, M.; Chieco, P.; Negrini, M.; Calin, G.A.; Grazi, G.L.; Pollutri, D.; Croce, C.M.; Bolondi, L.; Gramantieri, L.; et al. MiR-199a-3p Regulates mTOR and c-Met to Influence the Doxorubicin Sensitivity of Human Hepatocarcinoma Cells. Cancer Res. 2010, 70, 5184–5193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Hou, J.; Lin, L.; Zhou, W.; Wang, Z.; Ding, G.; Dong, Q.; Qin, L.; Wu, X.; Zheng, Y.; Yang, Y.; et al. Identification of miRNomes in Human Liver and Hepatocellular Carcinoma Reveals miR-199a/b-3p as Therapeutic Target for Hepatocellular Carcinoma. Cancer Cell 2011, 19, 232–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Shatseva, T.; Lee, D.Y.; Deng, Z.; Yang, B.B. MicroRNA miR-199a-3p regulates cell proliferation and survival by targeting caveolin-2. J. Cell Sci. 2011, 124, 2826–2836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Shen, Q.; Cicinnati, V.R.; Zhang, X.; Iacob, S.; Weber, F.; Sotiropoulos, G.C.; Radtke, A.; Lu, M.; Paul, A.; Gerken, G.; et al. Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol. Cancer 2010, 9, 227. [Google Scholar] [CrossRef] [Green Version]
  195. Xu, N.; Zhang, J.; Shen, C.; Luo, Y.; Xia, L.; Xue, F.; Xia, Q. Cisplatin-induced downregulation of miR-199a-5p increases drug resistance by activating autophagy in HCC cell. Biochem. Biophys. Res. Commun. 2012, 423, 826–831. [Google Scholar] [CrossRef]
  196. Gao, P.; Wong, C.C.-L.; Tung, E.K.-K.; Lee, J.M.-F.; Wong, C.-M.; Ng, I.O.L. Deregulation of microRNA expression occurs early and accumulates in early stages of HBV-associated multistep hepatocarcinogenesis. J. Hepatol. 2011, 54, 1177–1184. [Google Scholar] [CrossRef]
  197. Yuan, J.-H.; Yang, F.; Chen, B.-F.; Lu, Z.; Huo, X.-S.; Zhou, W.-P.; Wang, F.; Sun, S. The histone deacetylase 4/SP1/microrna-200a regulatory network contributes to aberrant histone acetylation in hepatocellular carcinoma. Hepatology 2011, 54, 2025–2035. [Google Scholar] [CrossRef]
  198. Wei, W.; Wanjun, L.; Hui, S.; Dongyue, C.; Xinjun, Y.; Jisheng, Z. miR-203 inhibits proliferation of HCC cells by targeting survivin. Cell Biochem. Funct. 2013, 31, 82–85. [Google Scholar] [CrossRef]
  199. Zhang, Y.; Wei, Y.; Li, X.; Liang, X.; Wang, L.; Song, J.; Zhang, X.; Zhang, C.; Niu, J.; Zhang, P.; et al. microRNA-874 suppresses tumor proliferation and metastasis in hepatocellular carcinoma by targeting the DOR/EGFR/ERK pathway. Cell Death Dis. 2018, 9, 130. [Google Scholar] [CrossRef]
  200. Jia, P.; Wei, G.; Zhou, C.; Gao, Q.; Wu, Y.; Sun, X.; Li, X. Upregulation of MiR-212 Inhibits Migration and Tumorigenicity and Inactivates Wnt/β-Catenin Signaling in Human Hepatocellular Carcinoma. Technol. Cancer Res. Treat. 2018, 17, 1533034618765221. [Google Scholar] [CrossRef] [Green Version]
  201. Shih, T.-C.; Tien, Y.-J.; Wen, C.-J.; Yeh, T.-S.; Yu, M.-C.; Huang, C.-H.; Lee, Y.-S.; Yen, T.-C.; Hsieh, S.-Y. MicroRNA-214 downregulation contributes to tumor angiogenesis by inducing secretion of the hepatoma-derived growth factor in human hepatoma. J. Hepatol. 2012, 57, 584–591. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, X.; Chen, J.; Li, F.; Lin, Y.; Zhang, X.; Lv, Z.; Jiang, J. MiR-214 inhibits cell growth in hepatocellular carcinoma through suppression of β-catenin. Biochem. Biophys. Res. Commun. 2012, 428, 525–531. [Google Scholar] [CrossRef] [PubMed]
  203. Xia, H.; Ooi, L.L.P.J.; Hui, K.M. MiR-214 Targets β-Catenin Pathway to Suppress Invasion, Stem-Like Traits and Recurrence of Human Hepatocellular Carcinoma. PLoS ONE 2012, 7, e44206. [Google Scholar] [CrossRef]
  204. Huang, N.; Lin, J.; Ruan, J.; Su, N.; Qing, R.; Liu, F.; He, B.; Lv, C.; Zheng, D.; Luo, R. MiR-219-5p inhibits hepatocellular carcinoma cell proliferation by targeting glypican-3. FEBS Lett. 2012, 586, 884–891. [Google Scholar] [CrossRef] [Green Version]
  205. Guichard, C.; Amaddeo, G.; Imbeaud, S.; Ladeiro, Y.; Pelletier, L.; Maad, I.B.; Calderaro, J.; Bioulac-Sage, P.; Letexier, M.; Degos, F.; et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 2012, 44, 694–698. [Google Scholar] [CrossRef]
  206. Wang, L.; Bo, X.; Zheng, Q.; Xiao, X.; Wu, L.; Li, B. miR-296 inhibits proliferation and induces apoptosis by targeting FGFR1 in human hepatocellular carcinoma. FEBS Lett. 2016, 590, 4252–4262. [Google Scholar] [CrossRef]
  207. Wang, L.; Yao, J.; Shi, X.; Hu, L.; Li, Z.; Song, T.; Huang, C. MicroRNA-302b suppresses cell proliferation by targeting EGFR in human hepatocellular carcinoma SMMC-7721 cells. BMC Cancer 2013, 13, 448. [Google Scholar] [CrossRef] [Green Version]
  208. Wang, L.; Yao, J.; Sun, H.; Sun, R.; Chang, S.; Yang, Y.; Song, T.; Huang, C. miR-302b suppresses cell invasion and metastasis by directly targeting AKT2 in human hepatocellular carcinoma cells. Tumor Biol. 2016, 37, 847–855. [Google Scholar] [CrossRef]
  209. Cui, H.; Song, R.; Wu, J.; Wang, W.; Chen, X.; Yin, J. MicroRNA-337 regulates the PI3K/AKT and Wnt/beta-catenin signaling pathways to inhibit hepatocellular carcinoma progression by targeting high-mobility group AT-hook 2. Am. J. Cancer Res. 2018, 8, 405–421. [Google Scholar]
  210. Zhang, T.; Liu, W.; Zeng, X.-C.; Jiang, N.; Fu, B.-S.; Guo, Y.; Yi, H.; Li, H.; Zhang, Q.; Chen, W.-J.; et al. Down-regulation of microRNA-338-3p promoted angiogenesis in hepatocellular carcinoma. Biomed. Pharmacother. 2016, 84, 583–591. [Google Scholar] [CrossRef]
  211. Yuan, J.; Ji, H.; Xiao, F.; Lin, Z.; Zhao, X.; Wang, Z.; Zhao, J.; Lu, J. MicroRNA-340 inhibits the proliferation and invasion of hepatocellular carcinoma cells by targeting JAK1. Biochem. Biophys. Res. Commun. 2017, 483, 578–584. [Google Scholar] [CrossRef] [PubMed]
  212. Yu, M.; Xue, H.; Wang, Y.; Shen, Q.; Jiang, Q.; Zhang, X.; Li, K.; Jia, M.; Jia, J.; Xu, J.; et al. miR-345 inhibits tumor metastasis and EMT by targeting IRF1-mediated mTOR/STAT3/AKT pathway in hepatocellular carcinoma. Int. J. Oncol. 2017, 50, 975–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Pan, X.-P.; Wang, H.-X.; Tong, D.-M.; Li, Y.; Huang, L.-H.; Wang, C. miRNA-370 acts as a tumor suppressor via the downregulation of PIM1 in hepatocellular carcinoma. Eur. Rev. Med Pharmacol. Sci. 2017, 21, 1254–1263. [Google Scholar]
  214. Chang, Y.; Yan, W.; He, X.; Zhang, L.; Li, C.; Huang, H.; Nace, G.; Geller, D.A.; Lin, J.-S.; Tsung, A. miR-375 Inhibits Autophagy and Reduces Viability of Hepatocellular Carcinoma Cells Under Hypoxic Conditions. Gastroenterology 2012, 143, 177–187.e8. [Google Scholar] [CrossRef] [PubMed]
  215. He, X.-X.; Chang, Y.; Meng, F.-Y.; Wang, M.-Y.; Xie, Q.-H.; Tang, F.; Li, P.-Y.; Song, Y.-H.; Lin, J.-S. MicroRNA-375 targets AEG-1 in hepatocellular carcinoma and suppresses liver cancer cell growth in vitro and in vivo. Oncogene 2012, 31, 3357–3369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Zheng, Y.; Yin, L.; Chen, H.; Yang, S.; Pan, C.; Lu, S.; Miao, M.; Jiao, B. miR-376a suppresses proliferation and induces apoptosis in hepatocellular carcinoma. FEBS Lett. 2012, 586, 2396–2403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. You, X.; Liu, F.; Zhang, T.; Li, Y.; Ye, L.; Zhang, X. Hepatitis B virus X protein upregulates oncogene Rab18 to result in the dysregulation of lipogenesis and proliferation of hepatoma cells. Carcinogenesis 2013, 34, 1644–1652. [Google Scholar] [CrossRef] [Green Version]
  218. Buurman, R.; Gürlevik, E.; Schäffer, V.; Eilers, M.; Sandbothe, M.; Kreipe, H.; Wilkens, L.; Schlegelberger, B.; Kühnel, F.; Skawran, B. Histone Deacetylases Activate Hepatocyte Growth Factor Signaling by Repressing MicroRNA-449 in Hepatocellular Carcinoma Cells. Gastroenterology 2012, 143, 811–820.e15. [Google Scholar] [CrossRef]
  219. Weng, Z.; Wang, N.; Zhao, W.; Song, M.; You, F.; Yang, L.; Chen, L.-B. microRNA-450a targets DNA methyltransferase 3a in hepatocellular carcinoma. Exp. Ther. Med. 2011, 2, 951–955. [Google Scholar] [CrossRef] [Green Version]
  220. Liu, X.; Zhang, A.; Xiang, J.; Lv, Y.; Zhang, X. miR-451 acts as a suppressor of angiogenesis in hepatocellular carcinoma by targeting the IL-6R-STAT3 pathway. Oncol. Rep. 2016, 36, 1385–1392. [Google Scholar] [CrossRef]
  221. Ye, Y.; Zhuang, J.; Wang, G.; He, S.; Zhang, S.; Wang, G.; Ni, J.; Wang, J.; Xia, W. MicroRNA-495 suppresses cell proliferation and invasion of hepatocellular carcinoma by directly targeting insulin-like growth factor receptor-1. Exp. Ther. Med. 2018, 15, 1150–1158. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, M.; Wu, J.; Zhang, R.; Yang, J.; Zhang, Q.; Liu, B. miR-497 inhibits the carcinogenesis of hepatocellular carcinoma by targeting the Rictor/Akt signal pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 1992–2000. [Google Scholar]
  223. Zhang, W.; Kong, G.; Zhang, J.; Wang, T.; Ye, L.; Zhang, X. MicroRNA-520b Inhibits Growth of Hepatoma Cells by Targeting MEKK2 and Cyclin D1. PLoS ONE 2012, 7, e31450. [Google Scholar] [CrossRef] [Green Version]
  224. Zhang, S.; Shan, C.; Kong, G.; Du, Y.; Ye, L.; Zhang, X. MicroRNA-520e suppresses growth of hepatoma cells by targeting the NF-κB-inducing kinase (NIK). Oncogene 2011, 31, 3607–3620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Liu, Y.; Hong, W.; Zhou, C.; Jiang, Z.; Wang, G.; Wei, G.; Li, X. miR-539 inhibits FSCN1 expression and suppresses hepatocellular carcinoma migration and invasion. Oncol. Rep. 2017, 37, 2593–2602. [Google Scholar] [CrossRef] [PubMed]
  226. Tao, Z.-H.; Wan, J.-L.; Zeng, L.-Y.; Xie, L.; Sun, H.-C.; Qin, L.-X.; Wang, L.; Zhou, J.; Ren, Z.-G.; Li, Y.-X.; et al. miR-612 suppresses the invasive-metastatic cascade in hepatocellular carcinoma. J. Exp. Med. 2013, 210, 789–803. [Google Scholar] [CrossRef] [Green Version]
  227. Zhang, J.-F.; He, M.; Fu, W.-M.; Wang, H.; Chen, L.-Z.; Zhu, X.; Chen, Y.; Xie, D.; Lai, P.B.S.; Chen, G.; et al. Primate-specific microRNA-637 inhibits tumorigenesis in hepatocellular carcinoma by disrupting signal transducer and activator of transcription 3 signaling. Hepatology 2011, 54, 2137–2148. [Google Scholar] [CrossRef]
  228. Zhang, Y.; Zhang, D.; Jiang, J.; Dong, L. Loss of miR-638 promotes invasion and epithelial-mesenchymal transition by targeting SOX2 in hepatocellular carcinoma. Oncol. Rep. 2017, 37, 323–332. [Google Scholar] [CrossRef] [Green Version]
  229. Huang, W.; Li, J.; Guo, X.; Zhao, Y.; Yuan, X. miR-663a inhibits hepatocellular carcinoma cell proliferation and invasion by targeting HMGA2. Biomed. Pharmacother. 2016, 81, 431–438. [Google Scholar] [CrossRef]
  230. Ding, D.; Zhang, Y.; Yang, R.; Wang, X.; Ji, G.; Huo, L.; Shao, Z.; Li, X. miR-940 Suppresses Tumor Cell Invasion and Migration via Regulation of CXCR2 in Hepatocellular Carcinoma. BioMed Res. Int. 2016, 2016, 7618342. [Google Scholar] [CrossRef] [Green Version]
  231. Zhu, H.; Wang, G.; Zhou, X.; Song, X.; Gao, H.; Ma, C.; Chang, H.; Li, H.; Liu, F.-F.; Lu, J.; et al. miR-1299 suppresses cell proliferation of hepatocellular carcinoma (HCC) by targeting CDK6. Biomed. Pharmacother. 2016, 83, 792–797. [Google Scholar] [CrossRef] [PubMed]
  232. Yang, C.; Xu, Y.; Cheng, F.; Hu, Y.; Yang, S.; Rao, J.; Wang, X. miR-1301 inhibits hepatocellular carcinoma cell migration, invasion, and angiogenesis by decreasing Wnt/β-catenin signaling through targeting BCL9. Cell Death Dis. 2017, 8, e2999. [Google Scholar] [CrossRef] [PubMed]
  233. Li, Q.-J.; Zhou, L.; Yang, F.; Wang, G.-X.; Zheng, H.; Wang, D.-S.; He, Y.; Dou, K.-F. MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells. Tumor Biol. 2012, 33, 1455–1465. [Google Scholar] [CrossRef] [PubMed]
  234. Zhu, Q.; Gong, L.; Wang, J.; Tu, Q.; Yao, L.; Zhang, J.-R.; Han, X.-J.; Zhu, S.-J.; Wang, S.-M.; Li, Y.-H.; et al. miR-10b exerts oncogenic activity in human hepatocellular carcinoma cells by targeting expression of CUB and sushi multiple domains 1 (CSMD1). BMC Cancer 2016, 16, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Yang, F.; Yin, Y.; Wang, F.; Wang, Y.; Zhang, L.; Tang, Y.; Sun, S. miR-17-5p Promotes migration of human hepatocellular carcinoma cells through the p38 mitogen-activated protein kinase-heat shock protein 27 pathway. Hepatol. 2010, 51, 1614–1623. [Google Scholar] [CrossRef] [PubMed]
  236. Liu, W.; Yeh, S.; Lu, C.; Yu, S.; Chen, H.; Lin, C.; Chen, D.; Chen, P. MicroRNA-18a Prevents Estrogen Receptor-α Expression, Promoting Proliferation of Hepatocellular Carcinoma Cells. Gastroenterology 2009, 136, 683–693. [Google Scholar] [CrossRef]
  237. Jiang, R.; Deng, L.; Zhao, L.; Li, X.; Zhang, F.; Xia, Y.; Gao, Y.; Wang, X.; Sun, B. miR-22 Promotes HBV-Related Hepatocellular Carcinoma Development in Males. Clin. Cancer Res. 2011, 17, 5593–5603. [Google Scholar] [CrossRef] [Green Version]
  238. Yao, J.; Liang, L.; Huang, S.; Ding, J.; Tan, N.; Zhao, Y.; Yan, M.; Ge, C.; Zhang, Z.; Chen, T.; et al. MicroRNA-30d promotes tumor invasion and metastasis by targeting Galphai2 in hepatocellular carcinoma. Hepatology 2010, 51, 846–856. [Google Scholar] [CrossRef]
  239. Shen, G.; Jia, H.; Tai, Q.; Li, Y.; Chen, D. miR-106b downregulates adenomatous polyposis coli and promotes cell proliferation in human hepatocellular carcinoma. Carcinogenesis 2013, 34, 211–219. [Google Scholar] [CrossRef] [Green Version]
  240. Wang, Y.; Zhang, C.; Zhang, P.; Guo, G.; Jiang, T.; Zhao, X.; Jiang, J.; Huang, X.; Tong, H.; Tian, Y. Serum exosomal microRNAs combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer Med. 2018, 7, 1670–1679. [Google Scholar] [CrossRef] [Green Version]
  241. Ma, S.; Tang, K.H.; Chan, Y.P.; Lee, T.K.; Kwan, P.S.; Castilho, A.; Ng, I.; Man, K.; Wong, N.; To, K.F.; et al. miR-130b Promotes CD133(+) Liver Tumor-Initiating Cell Growth and Self-Renewal via Tumor Protein 53-Induced Nuclear Protein 1. Cell Stem Cell 2010, 7, 694–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Liu, S.; Guo, W.; Shi, J.; Li, N.; Yu, X.; Xue, J.; Fu, X.; Chu, K.; Lu, C.; Zhao, J.; et al. MicroRNA-135a contributes to the development of portal vein tumor thrombus by promoting metastasis in hepatocellular carcinoma. J. Hepatol. 2012, 56, 389–396. [Google Scholar] [CrossRef] [PubMed]
  243. Zhang, X.; Liu, S.; Hu, T.; Liu, S.; He, Y.; Sun, S. Up-regulated microRNA-143 transcribed by nuclear factor kappa B enhances hepatocarcinoma metastasis by repressing fibronectin expression. Hepatology 2009, 50, 490–499. [Google Scholar] [CrossRef] [PubMed]
  244. Luedde, T. MicroRNA-151 and its hosting gene FAK (focal adhesion kinase) regulate tumor cell migration and spreading of hepatocellular carcinoma. Hepatology 2010, 52, 1164–1166. [Google Scholar] [CrossRef] [PubMed]
  245. Ding, J.; Huang, S.; Wu, S.; Zhao, Y.; Liang, L.; Yan, M.; Ge, C.; Yao, J.; Chen, T.; Wan, D.; et al. Gain of miR-151 on chromosome 8q24.3 facilitates tumour cell migration and spreading through downregulating RhoGDIA. Nat. Cell Biol. 2010, 12, 390–399. [Google Scholar] [CrossRef] [PubMed]
  246. Zhang, Y.; Wei, W.; Cheng, N.; Wang, K.; Li, B.; Jiang, X.; Sun, S. Hepatitis C virus-induced up-regulation of microRNA-155 promotes hepatocarcinogenesis by activating Wnt signaling. Hepatology 2012, 56, 1631–1640. [Google Scholar] [CrossRef]
  247. Yan, X.-L.; Jia, Y.-L.; Chen, L.; Zeng, Q.; Zhou, J.-N.; Fu, C.-J.; Chen, H.-X.; Yuan, H.-F.; Li, Z.-W.; Shi, L.; et al. Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression: Role of the S100A4-miR155-SOCS1-MMP9 axis. Hepatology 2013, 57, 2274–2286. [Google Scholar] [CrossRef]
  248. Wang, B.; Hsu, S.-H.; Majumder, S.K.; Kutay, H.; Huang, W.; Jacob, S.T.; Ghoshal, K. TGFβ-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene 2010, 29, 1787–1797. [Google Scholar] [CrossRef] [Green Version]
  249. Wang, J.; Li, J.; Shen, J.; Wang, C.; Yang, L.; Zhang, X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer 2012, 12, 227. [Google Scholar] [CrossRef] [Green Version]
  250. Goeppert, B.; Schmezer, P.; Dutruel, C.; Oakes, C.; Renner, M.; Breinig, M.; Warth, A.; Vogel, M.N.; Mittelbronn, M.; Mehrabi, A.; et al. Down-regulation of tumor suppressor a kinase anchor protein 12 in human hepatocarcinogenesis by epigenetic mechanisms. Hepatology 2010, 52, 2023–2033. [Google Scholar] [CrossRef]
  251. Petrelli, A.; Perra, A.; Cora, D.; Sulas, P.; Menegon, S.; Manca, C.; Migliore, C.; Kowalik, M.A.; Ledda-Columbano, G.M.; Giordano, S.; et al. MicroRNA/gene profiling unveils early molecular changes and nuclear factor erythroid related factor 2 (NRF2) activation in a rat model recapitulating human hepatocellular carcinoma (HCC). Hepatology 2014, 59, 228–241. [Google Scholar] [CrossRef] [PubMed]
  252. Fu, X.; Wen, H.; Jing, L.; Yang, Y.; Wang, W.; Liang, X.; Nan, K.; Yao, Y.; Tian, T. MicroRNA-155-5p promotes hepatocellular carcinoma progression by suppressing PTEN through the PI3K/Akt pathway. Cancer Sci. 2017, 108, 620–631. [Google Scholar] [CrossRef] [PubMed]
  253. Ying, Q.; Liang, L.; Guo, W.; Zha, R.; Tian, Q.; Huang, S.; Yao, J.; Ding, J.; Bao, M.; Ge, C.; et al. Hypoxia-inducible MicroRNA-210 augments the metastatic potential of tumor cells by targeting vacuole membrane protein 1 in hepatocellular carcinoma. Hepatology 2011, 54, 2064–2075. [Google Scholar] [CrossRef] [PubMed]
  254. Li, H.; Wang, H.; Ren, Z. MicroRNA-214-5p Inhibits the Invasion and Migration of Hepatocellular Carcinoma Cells by Targeting Wiskott-Aldrich Syndrome Like. Cell. Physiol. Biochem. 2018, 46, 757–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Chen, P.-J.; Yeh, S.; Liu, W.-H.; Lin, C.-C.; Huang, H.-C.; Chen, C.-L.; Chen, D.-S.; Chen, P.-J. Androgen pathway stimulates MicroRNA-216a transcription to suppress the tumor suppressor in lung cancer-1 gene in early hepatocarcinogenesis. Hepatology 2012, 56, 632–643. [Google Scholar] [CrossRef] [PubMed]
  256. Xia, H.; Ooi, L.L.P.; Hui, K.M. MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology 2013, 58, 629–641. [Google Scholar] [CrossRef]
  257. Callegari, E.; Elamin, B.K.; Giannone, F.; Milazzo, M.; Altavilla, G.; Fornari, F.; Giacomelli, L.; D’Abundo, L.; Ferracin, M.; Bassi, C.; et al. Liver tumorigenicity promoted by microRNA-221 in a mouse transgenic model. Hepatology 2012, 56, 1025–1033. [Google Scholar] [CrossRef]
  258. Yuan, Q.; Loya, K.; Rani, B.; Möbus, S.; Balakrishnan, A.; Lamle, J.; Cathomen, T.; Vogel, A.; Manns, M.P.; Ott, M.; et al. MicroRNA-221 overexpression accelerates hepatocyte proliferation during liver regeneration. Hepatology 2013, 57, 299–310. [Google Scholar] [CrossRef]
  259. Gramantieri, L.; Fornari, F.; Ferracin, M.; Veronese, A.; Sabbioni, S.; Calin, G.A.; Grazi, G.L.; Croce, C.M.; Bolondi, L.; Negrini, M. MicroRNA-221 Targets Bmf in Hepatocellular Carcinoma and Correlates with Tumor Multifocality. Clin. Cancer Res. 2009, 15, 5073–5081. [Google Scholar] [CrossRef] [Green Version]
  260. Pineau, P.; Volinia, S.; McJunkin, K.; Marchio, A.; Battiston, C.; Terris, B.; Mazzaferro, V.; Lowe, S.W.; Croce, C.M.; Dejean, A. miR-221 overexpression contributes to liver tumorigenesis. Proc. Natl. Acad. Sci. USA 2010, 107, 264–269. [Google Scholar] [CrossRef] [Green Version]
  261. Lan, S.; Wu, S.; Zuchini, R.; Lin, X.; Su, I.; Tsai, T.; Lin, Y.; Wu, C.; Liu, H. Autophagy suppresses tumorigenesis of hepatitis B virus-associated hepatocellular carcinoma through degradation of microRNA-224. Hepatology 2014, 59, 505–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Scisciani, C.; Vossio, S.; Guerrieri, F.; Schinzari, V.; De Iaco, R.; de Meo, P.D.; Cervello, M.; Montalto, G.; Pollicino, T.; Raimondo, G.; et al. Transcriptional regulation of miR-224 upregulated in human HCCs by NFκB inflammatory pathways. J. Hepatol. 2012, 56, 855–861. [Google Scholar] [CrossRef] [PubMed]
  263. Zhou, P.; Jiang, W.; Wu, L.; Chang, R.; Wu, K.; Wang, Z. miR-301a Is a Candidate Oncogene that Targets the Homeobox Gene Gax in Human Hepatocellular Carcinoma. Dig. Dis. Sci. 2012, 57, 1171–1180. [Google Scholar] [CrossRef] [PubMed]
  264. Chen, Y.-L.; Xu, Q.-P.; Guo, F.; Guan, W.-H. MicroRNA-302d downregulates TGFBR2 expression and promotes hepatocellular carcinoma growth and invasion. Exp. Ther. Med. 2017, 13, 681–687. [Google Scholar] [CrossRef] [Green Version]
  265. Yu, Q.; Yang, X.; Duan, W.; Li, C.; Luo, Y.; Lu, S. miRNA-346 promotes proliferation, migration and invasion in liver cancer. Oncol. Lett. 2017, 14, 3255–3260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Wu, N.; Liu, X.; Xu, X.; Fan, X.; Liu, M.; Li, X.; Zhong, Q.; Tang, H. MicroRNA-373, a new regulator of protein phosphatase 6, functions as an oncogene in hepatocellular carcinoma. FEBS J. 2011, 278, 2044–2054. [Google Scholar] [CrossRef] [PubMed]
  267. Lin, J.; Huang, S.; Wu, S.; Ding, J.; Zhao, Y.; Liang, L.; Tian, Q.; Zha, R.; Zhan, R.; He, X. MicroRNA-423 promotes cell growth and regulates G 1 /S transition by targeting p21Cip1/Waf1 in hepatocellular carcinoma. Carcinogenesis 2011, 32, 1641–1647. [Google Scholar] [CrossRef]
  268. Yang, H.; Cho, M.E.; Li, T.W.; Peng, H.; Ko, K.S.; Mato, J.M.; Lu, S.C. MicroRNAs regulate methionine adenosyltransferase 1A expression in hepatocellular carcinoma. J. Clin. Investig. 2012, 123, 285–298. [Google Scholar] [CrossRef]
  269. Chang, R.-M.; Xiao, S.; Lei, X.; Yang, H.; Fang, F.; Yang, L.-Y. miRNA-487a Promotes Proliferation and Metastasis in Hepatocellular Carcinoma. Clin. Cancer Res. 2017, 23, 2593–2604. [Google Scholar] [CrossRef] [Green Version]
  270. Zhang, L.-Y.; Liu, M.; Li, X.; Tang, H. miR-490-3p Modulates Cell Growth and Epithelial to Mesenchymal Transition of Hepatocellular Carcinoma Cells by Targeting Endoplasmic Reticulum-Golgi Intermediate Compartment Protein 3 (ERGIC3). J. Biol. Chem. 2013, 288, 4035–4047. [Google Scholar] [CrossRef] [Green Version]
  271. Lim, L.; Balakrishnan, A.; Huskey, N.; Jones, K.D.; Jodari, M.; Ng, R.; Song, G.; Riordan, J.; Anderton, B.; Cheung, S.; et al. MicroRNA-494 within an oncogenic microRNA megacluster regulates G 1 /S transition in liver tumorigenesis through suppression of mutated in colorectal cancer. Hepatology 2014, 59, 202–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Toffanin, S.; Hoshida, Y.; Lachenmayer, A.; Villanueva, A.; Cabellos, L.; Minguez, B.; Savic, R.; Ward, S.C.; Thung, S.; Chiang, D.Y.; et al. MicroRNA-Based Classification of Hepatocellular Carcinoma and Oncogenic Role of miR-517a. Gastroenterology 2011, 140, 1618–1628.e16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Fornari, F.; Milazzo, M.; Chieco, P.; Negrini, M.; Marasco, E.; Capranico, G.; Mantovani, V.; Marinello, J.; Sabbioni, S.; Callegari, E.; et al. In hepatocellular carcinoma miR-519d is up-regulated by p53 and DNA hypomethylation and targets CDKN1A/p21, PTEN, AKT3 and TIMP2. J. Pathol. 2012, 227, 275–285. [Google Scholar] [CrossRef] [PubMed]
  274. Tian, Q.; Liang, L.; Ding, J.; Zha, R.; Shi, H.; Wang, Q.; Huang, S.; Guo, W.; Ge, C.; Chen, T.; et al. MicroRNA-550a Acts as a Pro-Metastatic Gene and Directly Targets Cytoplasmic Polyadenylation Element-Binding Protein 4 in Hepatocellular Carcinoma. PLoS ONE 2012, 7, e48958. [Google Scholar] [CrossRef] [PubMed]
  275. Jiang, X.; Xiang, G.; Wang, Y.; Zhang, L.; Yang, X.; Cao, L.; Peng, H.; Xue, P.; Chen, D. MicroRNA-590-5p regulates proliferation and invasion in human hepatocellular carcinoma cells by targeting TGF-β RII. Mol. Cells 2012, 33, 545–551. [Google Scholar] [CrossRef] [PubMed]
  276. El Tayebi, H.; Hosny, K.; Esmat, G.; Breuhahn, K.; Abdelaziz, A.I. miR-615-5p is restrictedly expressed in cirrhotic and cancerous liver tissues and its overexpression alleviates the tumorigenic effects in hepatocellular carcinoma. FEBS Lett. 2012, 586, 3309–3316. [Google Scholar] [CrossRef] [Green Version]
  277. Zhang, L.; Yang, L.X.; Liu, X.; Chen, W.; Chang, L.; Chen, L.; Loera, S.; Chu, P.; Huang, W.-C.; Liu, Y.-R.; et al. MicroRNA-657 promotes tumorigenesis in hepatocellular carcinoma by targeting transducin-like enhancer protein 1 through nuclear factor kappa B pathways. Hepatology 2013, 57, 1919–1930. [Google Scholar] [CrossRef]
  278. Xie, B.; Guo-Sheng, T.; Hua, R.-X.; Zhang, B.; Tan, G.; Xiong, S.; Liu, L.; Chen, W.; Yang, J.; Wang, X.-N.; et al. Mir-765 promotes cell proliferation by downregulating INPP4B expression in human hepatocellular carcinoma. Cancer Biomark. 2016, 16, 405–413. [Google Scholar] [CrossRef]
  279. Han, G.; Zhang, L.; Ni, X.; Chen, Z.; Pan, X.; Zhu, Q.; Li, S.; Wu, J.; Huang, X.; Wang, X. MicroRNA-873 Promotes Cell Proliferation, Migration, and Invasion by Directly Targeting TSLC1 in Hepatocellular Carcinoma. Cell. Physiol. Biochem. 2018, 46, 2261–2270. [Google Scholar] [CrossRef] [Green Version]
  280. Jia, B.; Tan, L.; Jin, Z.; Jiao, Y.; Fu, Y.; Liu, Y. MiR-892a Promotes Hepatocellular Carcinoma Cells Proliferation and Invasion Through Targeting CD226. J. Cell. Biochem. 2017, 118, 1489–1496. [Google Scholar] [CrossRef]
  281. Ye, Y.; Wei, Y.; Xu, Y.; Li, Y.; Wang, R.; Chen, J.; Zhou, Y.; Fu, Z.; Chen, Y.; Wang, X.; et al. Induced MiR-1249 expression by aberrant activation of Hedegehog signaling pathway in hepatocellular carcinoma. Exp. Cell Res. 2017, 355, 9–17. [Google Scholar] [CrossRef] [PubMed]
  282. Law, P.T.-Y.; Qin, H.; Ching, A.K.; Lai, K.P.; Na Co, N.; He, M.; Lung, R.W.-M.; Chan, A.W.-H.; Chan, T.-F.; Wong, N. Deep sequencing of small RNA transcriptome reveals novel non-coding RNAs in hepatocellular carcinoma. J. Hepatol. 2013, 58, 1165–1173. [Google Scholar] [CrossRef] [PubMed]
  283. Liu, Z.; Wang, Y.; Dou, C.; Sun, L.; Li, Q.; Wang, L.; Xu, Q.; Yang, W.; Liu, Q.; Tu, K. MicroRNA-1468 promotes tumor progression by activating PPAR-γ-mediated AKT signaling in human hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2018, 37, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Cheng, L.; Wang, H.; Han, S.-Y. MiR-3910 Promotes the Growth and Migration of Cancer Cells in the Progression of Hepatocellular Carcinoma. Dig. Dis. Sci. 2017, 62, 2812–2820. [Google Scholar] [CrossRef]
  285. Song, L.J.; Zhang, W.; Chang, Z.; Pan, Y.; Zong, H.; Fan, Q.; Wang, L.X. miR-4417 Targets Tripartite Motif-Containing 35 (TRIM35) and Regulates Pyruvate Kinase Muscle 2 (PKM2) Phosphorylation to Promote Proliferation and Suppress Apoptosis in Hepatocellular Carcinoma Cells. Med Sci. Monit. 2017, 23, 1741–1750. [Google Scholar] [CrossRef] [Green Version]
  286. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  287. Mao, B.; Wang, G. MicroRNAs involved with hepatocellular carcinoma (Review). Oncol. Rep. 2015, 34, 2811–2820. [Google Scholar] [CrossRef]
  288. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  289. Zhang, T.; Yang, Z.; Kusumanchi, P.; Han, S.; Liangpunsakul, S. Critical Role of microRNA-21 in the Pathogenesis of Liver Diseases. Front. Med. 2020, 7, 7. [Google Scholar] [CrossRef] [Green Version]
  290. Wei, J.; Feng, L.; Li, Z.; Xu, G.; Fan, X. MicroRNA-21 activates hepatic stellate cells via PTEN/Akt signaling. Biomed. Pharmacother. 2013, 67, 387–392. [Google Scholar] [CrossRef]
  291. Marquez, R.T.; Bandyopadhyay, S.; Wendlandt, E.B.; Keck, K.; Hoffer, B.A.; Icardi, M.S.; Christensen, R.N.; Schmidt, W.N.; McCaffrey, A.P. Correlation between microRNA expression levels and clinical parameters associated with chronic hepatitis C viral infection in humans. Lab. Investig. 2010, 90, 1727–1736. [Google Scholar] [CrossRef] [Green Version]
  292. Chen, B.-L.; Peng, J.; Li, Q.-F.; Yang, M.; Wang, Y.; Chen, W. Exogenous bone morphogenetic protein-7 reduces hepatic fibrosis inSchistosoma japonicum-infected miceviatransforming growth factor-β/Smad signaling. World J. Gastroenterol. 2013, 19, 1405–1415. [Google Scholar] [CrossRef]
  293. Luo, X.; Zhang, D.; Xie, J.; Su, Q.; He, X.; Bai, R.; Gao, G.; Pan, W. MicroRNA-96 Promotes Schistosomiasis Hepatic Fibrosis in Mice by Suppressing Smad7. Mol. Ther. Methods Clin. Dev. 2018, 11, 73–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Guo, X.; Lv, X.; Lv, X.; Ma, Y.; Chen, L.; Chen, Y. Circulating miR-21 serves as a serum biomarker for hepatocellular carcinoma and correlated with distant metastasis. Oncotarget 2017, 8, 44050–44058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Yoon, J.S.; Kim, G.; Lee, Y.R.; Park, S.Y.; Tak, W.Y.; Kweon, Y.O.; Park, J.G.; Lee, H.W.; Han, Y.S.; Ha, H.T.; et al. Clinical significance of microRNA-21 expression in disease progression of patients with hepatocellular carcinoma. Biomark. Med. 2018, 12, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  296. Wang, J.; Chu, Y.; Xu, M.; Zhang, X.; Zhou, Y. miR-21 promotes cell migration and invasion of hepatocellular carcinoma by targeting KLF5. Oncol. Lett. 2019, 17, 2221–2227. [Google Scholar] [CrossRef] [Green Version]
  297. Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. 2018, 37, 324. [Google Scholar] [CrossRef] [Green Version]
  298. Momen-Heravi, F.; Bala, S.; Kodys, K.; Szabo, G. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci. Rep. 2015, 5, 9991. [Google Scholar] [CrossRef] [Green Version]
  299. Bala, S.; Petrasek, J.; Mundkur, S.; Catalano, D.; Levin, I.; Ward, J.; Alao, H.; Kodys, K.; Szabo, G. Circulating microRNAs in exosomes indicate hepatocyte injury and inflammation in alcoholic, drug-induced, and inflammatory liver diseases. Hepatology 2012, 56, 1946–1957. [Google Scholar] [CrossRef] [Green Version]
  300. Xiao, C.; Rajewsky, K. MicroRNA Control in the Immune System: Basic Principles. Cell 2009, 136, 26–36. [Google Scholar] [CrossRef] [Green Version]
  301. Blaya, D.; Aguilar-Bravo, B.; Hao, F.; Casacuberta-Serra, S.; Coll, M.; Perea, L.; Vallverdú, J.; Graupera, I.; Pose, E.; Llovet, L.; et al. Expression of microRNA-155 in inflammatory cells modulates liver injury. Hepatology 2018, 68, 691–706. [Google Scholar] [CrossRef] [Green Version]
  302. Li, F.; Hu, Q.; Pang, Z.; Xu, X. LncRNA MAGI2-AS3 Upregulates Cytokine Signaling 1 by Sponging miR-155 in Non-Small Cell Lung Cancer. Cancer Biother. Radiopharm. 2020, 35, 72–76. [Google Scholar] [CrossRef] [PubMed]
  303. Li, N.; Cui, T.; Guo, W.; Wang, D.; Mao, L. MiR-155-5p accelerates the metastasis of cervical cancer cell via targeting TP53INP1. OncoTargets Ther. 2019, 12, 3181–3196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Liu, W.; Zhao, Y.; Zhang, T.-P.; Zhou, L.; Cui, Q.-C.; Zhou, W.-X.; You, L.; Chen, G.; Shu, H. MLH1 as a Direct Target of MiR-155 and a Potential Predictor of Favorable Prognosis in Pancreatic Cancer. J. Gastrointest. Surg. 2013, 17, 1399–1405. [Google Scholar] [CrossRef] [PubMed]
  305. Chen, G.; Wang, D.; Zhao, X.; Cao, J.; Zhao, Y.; Wang, F.; Bai, J.; Luo, D.; Li, L. miR-155-5p modulates malignant behaviors of hepatocellular carcinoma by directly targeting CTHRC1 and indirectly regulating GSK-3β-involved Wnt/β-catenin signaling. Cancer Cell Int. 2017, 17, 118. [Google Scholar] [CrossRef] [Green Version]
  306. Yan, H.; Guo, M. Schizandrin A inhibits cellular phenotypes of breast cancer cells by repressing miR -155. IUBMB Life 2020, 72, 1640–1648. [Google Scholar] [CrossRef]
  307. Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [Green Version]
  308. Chen, Z.; Ma, T.; Huang, C.; Hu, T.; Li, J. The Pivotal Role of microRNA-155 in the Control of Cancer. J. Cell. Physiol. 2014, 229, 545–550. [Google Scholar] [CrossRef]
  309. Garofalo, M.; Di Leva, G.; Romano, G.; Nuovo, G.; Suh, S.-S.; Ngankeu, A.; Taccioli, C.; Pichiorri, F.; Alder, H.; Secchiero, P.; et al. miR-221&222 Regulate TRAIL Resistance and Enhance Tumorigenicity through PTEN and TIMP3 Downregulation. Cancer Cell 2009, 16, 498–509. [Google Scholar] [CrossRef] [Green Version]
  310. Galardi, S.; Mercatelli, N.; Giorda, E.; Massalini, S.; Frajese, G.V.; Ciafrè, S.A.; Farace, M.G. miR-221 and miR-222 Expression Affects the Proliferation Potential of Human Prostate Carcinoma Cell Lines by Targeting p27Kip1. J. Biol. Chem. 2007, 282, 23716–23724. [Google Scholar] [CrossRef] [Green Version]
  311. Le Sage, C.; Nagel, R.; Egan, D.A.; Schrier, M.; Mesman, E.; Mangiola, A.; Anile, C.; Maira, G.; Mercatelli, N.; Ciafrè, S.A.; et al. Regulation of the p27Kip1 tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J. 2007, 26, 3699–3708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Visone, R.; Russo, L.; Pallante, P.; De Martino, I.; Ferraro, A.; Leone, V.; Borbone, E.; Petrocca, F.; Alder, H.; Croce, C.M.; et al. MicroRNAs (miR)-221 and miR-222, both overexpressed in human thyroid papillary carcinomas, regulate p27Kip1 protein levels and cell cycle. Endocrine-Related Cancer 2007, 14, 791–798. [Google Scholar] [CrossRef] [PubMed]
  313. Fornari, F.A.; Gramantieri, L.; Ferracin, M.; Veronese, A.; Sabbioni, S.; Calin, G.A.; Grazi, G.L.; Giovannini, C.; Croce, C.M.; Bolondi, L.; et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene 2008, 27, 5651–5661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Medina, R.F.; Zaidi, S.K.; Liu, C.-G.; Stein, J.L.; Van Wijnen, A.J.; Croce, C.M.; Stein, G.S. MicroRNAs 221 and 222 Bypass Quiescence and Compromise Cell Survival. Cancer Res. 2008, 68, 2773–2780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Yuan, B.; Dong, R.; Shi, D.; Zhou, Y.; Zhao, Y.; Miao, M.; Jiao, B. Down-regulation of miR-23b may contribute to activation of the TGF-β1/Smad3 signalling pathway during the termination stage of liver regeneration. FEBS Lett. 2011, 585, 927–934. [Google Scholar] [CrossRef] [Green Version]
  316. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. RAS Is Regulated by the let-7 MicroRNA Family. Cell 2005, 120, 635–647. [Google Scholar] [CrossRef] [Green Version]
  317. Matsuura, K.; De Giorgi, V.; Schechterly, C.; Wang, R.Y.; Farci, P.; Tanaka, Y.; Alter, H.J. Circulating let-7 levels in plasma and extracellular vesicles correlate with hepatic fibrosis progression in chronic hepatitis C. Hepatology 2016, 64, 732–745. [Google Scholar] [CrossRef]
  318. Winkler, I.; Bitter, C.; Winkler, S.; Weichenhan, D.; Thavamani, A.; Hengstler, J.G.; Borkham-Kamphorst, E.; Kohlbacher, O.; Plass, C.; Geffers, R.; et al. Identification of Pparγ-modulated miRNA hubs that target the fibrotic tumor microenvironment. Proc. Natl. Acad. Sci. USA 2020, 117, 454–463. [Google Scholar] [CrossRef] [Green Version]
  319. Ding, J.; Huang, S.; Wang, Y.; Tian, Q.; Zha, R.; Shi, H.; Wang, Q.; Ge, C.; Chen, T.; Zhao, Y.; et al. Genome-wide screening reveals that miR-195 targets the TNF-α/NF-κB pathway by down-regulating IκB kinase alpha and TAB3 in hepatocellular carcinoma. Hepatology 2013, 58, 654–666. [Google Scholar] [CrossRef]
  320. Wang, R.; Zhao, N.; Li, S.; Fang, J.-H.; Chen, M.-X.; Yang, J.-E.; Jia, W.-H.; Yuan, Y.; Zhuang, S.-M. MicroRNA-195 Suppresses Angiogenesis and Metastasis of Hepatocellular Carcinoma by Inhibiting the Expression of VEGF, VAV2, and CDC42. Hepatology 2013, 58, 642–653. [Google Scholar] [CrossRef]
  321. Kwon, J.J.; Factora, T.D.; Dey, S.; Kota, J. A Systematic Review of miR-29 in Cancer. Mol. Ther. Oncolytics 2019, 12, 173–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  322. Xiong, Y.; Fang, J.-H.; Yun, J.-P.; Yang, J.; Zhang, Y.; Jia, W.-H.; Zhuang, S.-M. Effects of MicroRNA-29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology 2010, 51, 836–845. [Google Scholar] [CrossRef] [PubMed]
  323. Weidle, U.H.; Schmid, D.; Birzele, F.; Brinkmann, U. MicroRNAs Involved in Metastasis of Hepatocellular Carcinoma: Target Candidates, Functionality and Efficacy in Animal Models and Prognostic Relevance. Cancer Genom. Proteom. 2020, 17, 1–21. [Google Scholar] [CrossRef] [PubMed]
  324. Cheng, D.; Deng, J.; Zhang, B.; He, X.; Meng, Z.; Li, G.; Ye, H.; Zheng, S.; Wei, L.; Deng, X.; et al. LncRNA HOTAIR epigenetically suppresses miR-122 expression in hepatocellular carcinoma via DNA methylation. EBioMedicine 2018, 36, 159–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Biselli-Chicote, P.M.; Oliveira, A.R.C.P.; Pavarino, E.C.; Goloni-Bertollo, E.M. VEGF gene alternative splicing: Pro- and anti-angiogenic isoforms in cancer. J. Cancer Res. Clin. Oncol. 2011, 138, 363–370. [Google Scholar] [CrossRef] [PubMed]
  326. Bupathi, M.; Kaseb, A.O.; Meric-Bernstam, F.; Naing, A. Hepatocellular carcinoma: Where there is unmet need. Mol. Oncol. 2015, 9, 1501–1509. [Google Scholar] [CrossRef] [Green Version]
  327. Llovet, J.M. Focal Gains of VEGFA: Candidate Predictors of Sorafenib Response in Hepatocellular Carcinoma. Cancer Cell 2014, 25, 560–562. [Google Scholar] [CrossRef] [Green Version]
  328. De Oliveira, A.R.; Castanhole-Nunes, M.M.; Biselli-Chicote, P.M.; Pavarino, É.C.; da Silva, R.; Da Silva, R.F.; Goloni-Bertollo, E.M. Differential expression of angiogenesis-related miRNAs and VEGFA in cirrhosis and hepatocellular carcinoma. Arch. Med Sci. 2020, 16, 1150–1157. [Google Scholar] [CrossRef]
  329. Chen, L.; Ma, H.; Hu, H.; Gao, L.; Wang, X.; Ma, J.; Gao, Q.; Liu, B.; Zhou, G.; Liang, C. Special role of Foxp3 for the specifically altered microRNAs in Regulatory T cells of HCC patients. BMC Cancer 2014, 14, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  330. Li, A.; Shuai, X.; Jia, Z.; Li, H.; Liang, X.; Su, D.; Guo, W. Ganoderma lucidum polysaccharide extract inhibits hepatocellular carcinoma growth by downregulating regulatory T cells accumulation and function by inducing microRNA-125b. J. Transl. Med. 2015, 13, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Tomimaru, Y.; Eguchi, H.; Nagano, H.; Wada, H.; Kobayashi, S.; Marubashi, S.; Tanemura, M.; Tomokuni, A.; Takemasa, I.; Umeshita, K.; et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J. Hepatol. 2012, 56, 167–175. [Google Scholar] [CrossRef] [PubMed]
  332. Gibbings, D.J.; Ciaudo, C.; Erhardt, M.; Voinnet, O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat. Cell Biol. 2009, 11, 1143–1149. [Google Scholar] [CrossRef] [PubMed]
  333. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [Green Version]
  334. Zhang, J.; Yang, Y.; Yang, T.; Liu, Y.; Li, A.; Fu, S.; Wu, M.; Pan, Z.; Zhou, W. microRNA-22, downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br. J. Cancer 2010, 103, 1215–1220. [Google Scholar] [CrossRef] [Green Version]
  335. Ji, J.; Shi, J.; Budhu, A.; Yu, Z.; Forgues, M.; Roessler, S.; Ambs, S.; Chen, Y.; Meltzer, P.S.; Croce, C.M.; et al. MicroRNA Expression, Survival, and Response to Interferon in Liver Cancer. N. Engl. J. Med. 2009, 361, 1437–1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Coulouarn, C.; Factor, V.M.; Andersen, J.B.; Durkin, M.E.; Thorgeirsson, S.S. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene 2009, 28, 3526–3536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Zheng, F.; Liao, Y.-J.; Cai, M.-Y.; Liu, Y.-H.; Liu, T.-H.; Chen, S.-P.; Bian, X.-W.; Guan, X.-Y.; Lin, M.C.-M.; Zeng, Y.-X.; et al. The putative tumour suppressor microRNA-124 modulates hepatocellular carcinoma cell aggressiveness by repressing ROCK2 and EZH2. Gut 2012, 61, 278–289. [Google Scholar] [CrossRef] [PubMed]
  338. Wong, C.C.; Wong, C.; Tung, E.K.; Au, S.L.; Lee, J.M.; Poon, R.T.; Man, K.; Ng, I.O. The MicroRNA miR-139 Suppresses Metastasis and Progression of Hepatocellular Carcinoma by Down-regulating Rho-Kinase 2. Gastroenterology 2011, 140, 322–331. [Google Scholar] [CrossRef]
  339. Wang, C.; Song, B.; Song, W.; Liu, J.; Sun, A.; Wu, D.; Yu, H.; Lian, J.; Chen, L.; Han, J. Underexpressed microRNA-199b-5p targets Hypoxia-Inducible Factor-1α in hepatocellular carcinoma and predicts prognosis of hepatocellular carcinoma patients. J. Gastroenterol. Hepatol. 2011, 26, 1630–1637. [Google Scholar] [CrossRef]
  340. Guo, Y.; Xiong, Y.; Sheng, Q.; Zhao, S.; Wattacheril, J.; Flynn, C.R. A micro-RNA expression signature for human NAFLD progression. J. Gastroenterol. 2016, 51, 1022–1030. [Google Scholar] [CrossRef] [Green Version]
  341. Chen, L.; Jiang, M.; Yuan, W.; Tang, H. miR-17-5p as a Novel Prognostic Marker for Hepatocellular Carcinoma. J. Investig. Surg. 2012, 25, 156–161. [Google Scholar] [CrossRef]
  342. Karakatsanis, A.; Papaconstantinou, I.; Gazouli, M.; Lyberopoulou, A.; Polymeneas, G.; Voros, D. Expression of microRNAs, miR-21, miR-31, miR-122, miR-145, miR-146a, miR-200c, miR-221, miR-222, and miR-223 in patients with hepatocellular carcinoma or intrahepatic cholangiocarcinoma and its prognostic significance. Mol. Carcinog. 2013, 52, 297–303. [Google Scholar] [CrossRef]
  343. Han, Z.-B.; Chen, H.; Fan, J.-W.; Wu, J.-Y.; Tang, H.-M.; Peng, Z.-H. Up-regulation of microRNA-155 promotes cancer cell invasion and predicts poor survival of hepatocellular carcinoma following liver transplantation. J. Cancer Res. Clin. Oncol. 2011, 138, 153–161. [Google Scholar] [CrossRef]
  344. Yamamoto, Y.; Kosaka, N.; Tanaka, M.; Koizumi, F.; Kanai, Y.; Mizutani, T.; Murakami, Y.; Kuroda, M.; Miyajima, A.; Kato, T.; et al. MicroRNA-500 as a potential diagnostic marker for hepatocellular carcinoma. Biomarkers 2009, 14, 529–538. [Google Scholar] [CrossRef] [PubMed]
  345. Li, L.-M.; Hu, Z.-B.; Zhou, Z.-X.; Chen, X.; Liu, F.-Y.; Zhang, J.-F.; Shen, H.-B.; Zhang, C.-Y.; Zen, K. Serum microRNA Profiles Serve as Novel Biomarkers for HBV Infection and Diagnosis of HBV-Positive Hepatocarcinoma. Cancer Res. 2010, 70, 9798–9807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  346. Xie, Y.; Yao, Q.; Butt, A.M.; Guo, J.; Tian, Z.; Bao, X.; Li, H.; Meng, Q.; Lu, J. Expression profiling of serum microRNA-101 in HBV-associated chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Cancer Biol. Ther. 2014, 15, 1248–1255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  347. Hung, C.-H.; Hu, T.-H.; Lu, S.-N.; Kuo, F.-Y.; Chen, C.-H.; Wang, J.-H.; Huang, C.-M.; Lee, C.-M.; Lin, C.-Y.; Yen, Y.-H.; et al. Circulating microRNAs as biomarkers for diagnosis of early hepatocellular carcinoma associated with hepatitis B virus. Int. J. Cancer 2016, 138, 714–720. [Google Scholar] [CrossRef] [PubMed]
  348. Zekri, A.-R.N.; Youssef, A.S.E.-D.; El-Desouky, E.D.; Ahmed, O.S.; Lotfy, M.M.; Nassar, A.A.-M.; Bahnassey, A.A. Serum microRNA panels as potential biomarkers for early detection of hepatocellular carcinoma on top of HCV infection. Tumor Biol. 2016, 37, 12273–12286. [Google Scholar] [CrossRef]
  349. Giray, B.G.; Emekdas, G.; Tezcan, S.; Ulger, M.; Serin, M.S.; Sezgin, O.; Altintas, E.; Tiftik, E.N. Profiles of serum microRNAs; miR-125b-5p and miR223-3p serve as novel biomarkers for HBV-positive hepatocellular carcinoma. Mol. Biol. Rep. 2014, 41, 4513–4519. [Google Scholar] [CrossRef]
  350. Fu, X.; Liu, M.; Qu, S.; Ma, J.; Zhang, Y.; Shi, T.; Wen, H.; Yang, Y.; Wang, S.; Wang, J.; et al. Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2018, 37, 52. [Google Scholar] [CrossRef] [Green Version]
  351. Chen, F.; Li, X.-F.; Fu, D.-S.; Huang, J.-G.; Yang, S.-E. Clinical potential of miRNA-221 as a novel prognostic biomarker for hepatocellular carcinoma. Cancer Biomark. 2017, 18, 209–214. [Google Scholar] [CrossRef] [PubMed]
  352. Shi, M.; Jiang, Y.; Yang, L.; Yan, S.; Wang, Y.-G.; Lu, X.-J. Decreased levels of serum exosomal miR-638 predict poor prognosis in hepatocellular carcinoma. J. Cell. Biochem. 2018, 119, 4711–4716. [Google Scholar] [CrossRef] [PubMed]
  353. Chung, H.J.; Choi, Y.E.; Kim, E.S.; Han, Y.-H.; Park, M.-J.; Bae, I.H. miR-29b attenuates tumorigenicity and stemness maintenance in human glioblastoma multiforme by directly targetingBCL2L2. Oncotarget 2015, 6, 18429–18444. [Google Scholar] [CrossRef] [PubMed]
  354. Kong, Y.W.; Ferland-McCollough, D.; Jackson, T.J.; Bushell, M. microRNAs in cancer management. Lancet Oncol. 2012, 13, e249–e258. [Google Scholar] [CrossRef]
  355. Amodeo, V.; Bazan, V.; Fanale, D.; Insalaco, L.; Caruso, S.; Cicero, G.; Bronte, G.; Rolfo, C.; Santini, D.; Russo, A. Effects of anti-miR-182 on TSP-1 expression in human colon cancer cells: There is a sense in antisense? Expert Opin. Ther. Targets 2013, 17, 1249–1261. [Google Scholar] [CrossRef] [Green Version]
  356. Nedaeinia, R.; Sharifi, M.; Avan, A.; Kazemi, M.; Rafiee, L.; Ghayour-Mobarhan, M.; Salehi, R. Locked nucleic acid anti-miR-21 inhibits cell growth and invasive behaviors of a colorectal adenocarcinoma cell line: LNA-anti-miR as a novel approach. Cancer Gene Ther. 2016, 23, 246–253. [Google Scholar] [CrossRef]
  357. Tsai, W.-C.; Hsu, P.W.-C.; Lai, T.-C.; Chau, G.-Y.; Lin, C.-W.; Chen, C.-M.; Lin, C.-D.; Liao, Y.-L.; Wang, J.-L.; Chau, Y.-P.; et al. MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology 2009, 49, 1571–1582. [Google Scholar] [CrossRef]
  358. Park, J.-K.; Kogure, T.; Nuovo, G.J.; Jiang, J.; He, L.; Kim, J.H.; Phelps, M.A.; Papenfuss, T.L.; Croce, C.M.; Patel, T.; et al. miR-221 Silencing Blocks Hepatocellular Carcinoma and Promotes Survival. Cancer Res. 2011, 71, 7608–7616. [Google Scholar] [CrossRef] [Green Version]
  359. Kato, K.; Gong, J.; Iwama, H.; Kitanaka, A.; Tani, J.; Miyoshi, H.; Nomura, K.; Mimura, S.; Kobayashi, M.; Aritomo, Y.; et al. The Antidiabetic Drug Metformin Inhibits Gastric Cancer Cell Proliferation In Vitro and In Vivo. Mol. Cancer Ther. 2012, 11, 549–560. [Google Scholar] [CrossRef] [Green Version]
  360. Kobayashi, M.; Kato, K.; Iwama, H.; Fujihara, S.; Nishiyama, N.; Mimura, S.; Toyota, Y.; Nomura, T.; Nomura, K.; Tani, J.; et al. Antitumor effect of metformin in esophageal cancer: In vitro study. Int. J. Oncol. 2013, 42, 517–524. [Google Scholar] [CrossRef] [Green Version]
  361. Miyoshi, H.; Kato, K.; Iwama, H.; Maeda, E.; Sakamoto, T.; Fujita, K.; Toyota, Y.; Tani, J.; Nomura, T.; Mimura, S.; et al. Effect of the anti-diabetic drug metformin in hepatocellular carcinoma in vitro and in vivo. Int. J. Oncol. 2014, 45, 322–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  362. Tomimaru, Y.; Eguchi, H.; Nagano, H.; Wada, H.; Tomokuni, A.; Kobayashi, S.; Marubashi, S.; Takeda, Y.; Tanemura, M.; Umeshita, K.; et al. MicroRNA-21 induces resistance to the anti-tumour effect of interferon-α/5-fluorouracil in hepatocellular carcinoma cells. Br. J. Cancer 2010, 103, 1617–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic of microRNA (miRNA, miR) biogenesis. (A) Synthesis of primary miRNA (pri-miRNA) transcripts from genomic DNA by RNA polymerase II. (B) The pri-miRNA is cleaved by Drosha/DiGeorge syndrome critical region 8 (DGCR8) and processed to a precursor miRNA (pre-miRNA). (C) The pre-miRNA forms a complex with exportin-5 and Ran-GTP and is exported to the cytoplasm. (D) The exported hairpin pre-miRNA is cleaved by Dicer/transactivation response element RNA-binding 70 protein (TRBP). (E) The double-stranded miRNA is unwound and forms an RNA-induced silencing complex (RISC) with Argonaute 2 (AGO2). (F) The miRNA is separated into a mature, single-stranded miRNA. (G) Upregulation of oncogenic miRs (oncomiRs) and downregulation of tumor suppressor miRs promote hepatocellular carcinoma (HCC) development.
Figure 1. Schematic of microRNA (miRNA, miR) biogenesis. (A) Synthesis of primary miRNA (pri-miRNA) transcripts from genomic DNA by RNA polymerase II. (B) The pri-miRNA is cleaved by Drosha/DiGeorge syndrome critical region 8 (DGCR8) and processed to a precursor miRNA (pre-miRNA). (C) The pre-miRNA forms a complex with exportin-5 and Ran-GTP and is exported to the cytoplasm. (D) The exported hairpin pre-miRNA is cleaved by Dicer/transactivation response element RNA-binding 70 protein (TRBP). (E) The double-stranded miRNA is unwound and forms an RNA-induced silencing complex (RISC) with Argonaute 2 (AGO2). (F) The miRNA is separated into a mature, single-stranded miRNA. (G) Upregulation of oncogenic miRs (oncomiRs) and downregulation of tumor suppressor miRs promote hepatocellular carcinoma (HCC) development.
Cancers 13 00514 g001
Table 1. Downregulated and upregulated miRNAs in HCC.
Table 1. Downregulated and upregulated miRNAs in HCC.
miRNATargetCategoryExpressionReferences
let-7aCaspase-3, STAT3apoptosis, proliferationDown[128,129,130]
let-7bHMGA2apoptosis, proliferationDown[131]
let-7cBcl-xL, c-mycapoptosis, proliferation, cell growthDown[132,133,134]
let-7d apoptosis, proliferationDown[129]
let-7f-1 apoptosis, proliferationDown[129]
let-7gBcl-xL, c-myc, COLIA2, p16 apoptosis, metastasisDown[135,136,137]
miR-1ET-1proliferationDown[138]
miR-7PIK3CD Down[139]
miR-10aEphA4 Down[140]
miR-10b Down[141]
miR-15a/16 Down[142]
miR-15bBCL-2proliferation, apoptosisDown[143]
miR-21 Down[144]
miR-26aCDK6, IL-6, cyclin D2, E1, E2Cell cycleDown[145,146,147]
miR-29aSPARCproliferationDown[148]
miR-29bMMP-2invasionDown[149]
miR-29cSIRT1 Down[150]
miR-31-5pSP1proliferation, migration, invasionDown[151]
miR-33bSALL4proliferation, metastasisDown[152]
miR-34ac-Met, CCL22metastasisDown[71,153]
miR-98EZH2proliferationDown[154]
miR-99aPLK1, IGF-1R Down[155,156]
miR-100PLK1carcinogenesisDown[155]
miR-101EZH2, EED, Mcl-1, Fos apoptosis, DNA methylationDown[157,158,159]
miR-122Bcl-w, ADAM17, Wnt1apoptosis, proliferation, angiogenesisDown[160,161,162,163]
miR-124PIK3CAproliferationDown[164]
miR-125aMMP11, VEGF-A, SIRT7proliferation, metastasis, metabolismDown[165,166]
miR-125bMcl-1, Bcl-w, SUV39H1, SIRT7proliferation, metastasis, angiogenesis, apoptosis, histone modificationDown[132,166,167,168,169,170]
miR-126VEGFangiogenesisDown[171]
miR-137EZH2proliferationDown[172]
miR-139ROCK2, c-FosmetastasisDown[132,173,174]
miR-139-5p Down[132]
miR-140-5pTGFβR1, FIF9, DMMT1 Down[175,176]
miR-141DLC-1 Down[95]
miR-142TGFβ, THBS4cell growth, metastasis, migration, invasionDown[177,178]
miR-142-3pLDHAproliferationDown[179]
miR-144ZFXproliferation, invasion, migrationDown[180]
miR-145IRS1/2, IGF-1R, β-catenin Down[181,182]
miR-148ac-Met, HRIP, E-cadherin, c-Myc Down[183,184,185,186]
miR-152DNMT1, GSTP1, CDH1 Down[187]
miR-187-3pS100A4metastasis, EMTDown[188]
miR-194MAP4K4proliferationDown[113]
miR-195cyclin D1, CDK6, E2F3, LATS2cell cycle, tumorigenesis, apoptosisDown[189,190]
miR-198 Down[39]
miR-199a-3pmTOR, PAK4, caveolin-2drug resistance, cell growthDown[191,192,193]
miR-199a-5pDDR1, ATG7invasion, autophagyDown[194,195]
miR-199b proliferation, invasion, apoptosisDown[196]
miR-200aHDAC4proliferation, metastasisDown[197]
miR-200b Down[132]
miR-200c Down[144]
miR-203SurvivingproliferationDown[198]
miR-206Cyclin D1, CDK6proliferationDown[199]
miR-212FOXM1migration, cell growthDown[200]
miR-214HDGF, β-catenincell growth, angiogenesis, metastasisDown[201,202,203]
miR-219-5pGPC3proliferationDown[204]
miR-222 Down[144]
miR-223STMN1proliferationDown[38]
miR-224 Down[205]
miR-296FGFR1proliferation, apoptosisDown[206]
miR-302bEGFR, AKT2proliferation, invasion, metastasisDown[207,208]
miR-337HMGA3proliferation, apoptosisDown[209]
miR-338-3pMACC1, β-catenin, VEGFangiogenesisDown[210]
miR-340JAK1proliferation, invasionDown[211]
miR-345IRF1metastasis, EMTDown[212]
miR-363-3pc-Myc Down[186]
miR-370PIM1cell growth, invasionDown[213]
miR-375ATG7, AEG-1autophagyDown[214,215]
miR-376aPIK3R1apoptosis, proliferationDown[216]
miR-429Rab18 Down[217]
miR-449c-Metproliferation, apoptosisDown[218]
miR-450aDNMT3aproliferationDown[219]
miR-451IL6RangiogenesisDown[220]
miR495IGF1Rproliferation, invasionDown[221]
miR-497RICTORproliferation, migration, invasionDown[222]
miR-520b/eNIK, MEKK2, cyclin D1cell growth, proliferationDown[223,224]
miR-539FSCN1migration, invasionDown[225]
miR-612AKT2 Down[226]
miR-637STAT activation Down[227]
miR-638SOX2, VEGFinvasion, EMT, angiogenesisDown[228]
miR-663aHMGA2proliferation, invasionDown[229]
miR-874DORproliferation, metastasisDown[199]
miR-940CXCR2migration, invasionDown[230]
miR-1271GLP3 Down[141]
miR-1299CDK6proliferationDown[231]
miR-1301BCL-9, β-catenin, VEGFAmigration, invasion, angiogenesisDown[232]
miR-10aEphA4; CADM1EMT, metastasisUp[140,233]
miR-10bCSMD 1division, migration, invasionUp[234]
miR-17-5pp38 pathwaymigrationUp[235]
miR-18aER1aproliferationUp[236]
miR-21PTEN; RHOB; PDCD4metastasis, drug resistanceUp[35,73]
miR-22Era, IL-1acarcinogenesisUp[237]
miR-23aPGC-1a, G6PCgluconeogenesisUp[50]
miR-25TRAILapoptosisUp[120]
miR-26aIL-6, CyclinD2, E2tumor growth, metastasisUp[145,146]
miR-30dGNAI2invasion, metastasisUp[238]
miR-92aFBXW 7Proliferation, cell cycle transition, apoptosisUp[125]
miR-96-5pCaspase-9apoptosisUp[121]
miR-100PLK1carcinogenesisUp[39]
miR-106bAPCproliferationUp[239]
miR-107HMGCS 2cell growth, EMTUp[123,240]
miR-130bTP53INP1cell growth, self-renewalUp[241]
miR-135aFOXM1, MTSS1metastasisUp[242]
miR-143FNDC3BmetastasisUp[243]
miR-151FAK, RhoGDIAmigrationUp[244,245]
miR-155SOCS1, DKK1, APC, PTENproliferation; TumorigenesisUp[73,246,247]
miR-181bTIMP3tumorigenesis, metastasisUp[248]
miR-182MTSS1metastasisUp[249]
miR-183AKAP12carcinogenesisUp[250]
miR-186AKAP12carcinogenesisUp[250]
miR-200NRF2 pathwaycarcinogenesisUp[251]
miR-203a-3pInterleukin (IL) 24cell growth, metastasisUp[252]
miR-210VMP1metastasisUp[253]
miR-214-5pWASLInvasion, migrationUp[254]
miR-216aTSLC1carcinogenesisUp[255]
miR-216a/217PTEN, SMAD7EMT, drug resistanceUp[256]
miR-221p27, p57, Arnt, CDK inhibitorsapoptosis, proliferation, angiogenesisUp[257,258,259]
miR-221/222p27, DDIT4tumorigenesisUp[260]
miR-224Atg5, Smad4, autophagy, API-5tumorigenesis, autophagyUp[261,262]
miR-301aGaxmetastasisUp[263]
miR-302dTGFβRIIcell growth, invasion,Up[264]
miR-346FBXL2proliferation, migration, invasionUp[265]
miR-373PPP6Ccell cycleUp[266]
miR-423p21/waf1cell growthUp[267]
miR-485-3pMAT1, LIN28Bcell growth, EMTUp[268]
miR-487aSPRED2, PIK3R1proliferation, metastasisUp[269]
miR-490-3pERCIC3EMTUp[270]
miR-494MCCtumorigenesisUp[271]
miR-495MAT1, LIN28Btumorigenesis, metastasisUp[268]
miR-517a tumorigenesis, metastasisUp[272]
miR-519dp21, PTEN, AKT3, TIMP2proliferation, invasion, apoptosisUp[273]
miR-550aCPEB4proliferation, invasion, metastasisUp[274]
miR-590-5pTGFβRIImetastasis, proliferationUp[275]
miR-615-5pIGF-IIproliferation, migrationUp[276]
miR-657TLE1, NF–κBproliferationUp[277]
miR-664MAT1, LIN28Btumorigenesis, metastasisUp[268]
miR-765 Up[278]
miR-873 Up[279]
miR-892a Up[280]
miR-1249 Up[281]
miR-1323 proliferationUp[282]
miR-1468 Up[283]
miR-3910 Up[284]
miR-4417 Up[285]
Table 2. miRNAs as biomarkers for HCC.
Table 2. miRNAs as biomarkers for HCC.
miRNAExpressionEtiologyReference
Diagnosislet-7gdownHBV[136]
miR-21upHBV, HCV and others[15,288]
miR-22downHBV[334]
miR-99adownHBV, HCV and others[156]
miR-101downHBV[346]
miR-122downHBV, HCV[347,348]
miR-124downHBV, HCV and others[337]
miR-125a-5pupHBV, HCV and others[349]
miR-135aupHBV, HCV and others[242]
miR-139downHBV and others[328]
miR-155upHBV[343]
miR-182upHBV and others[249]
miR-199bdownHBV, HCV and others[339]
miR-221upHBV, HCV and others[342]
miR-222upHBV, HCV and others[173,342]
miR-223-3pdownHBV[349]
miR-375upHBV[345]
Poor prognosismiR-32-5pupHBV and others[350]
miR-92aupHBV and others[125]
miR-122upHBV, HCV[240]
miR-137downHBV and others[172]
miR-148aupHBV, HCV[240]
miR-221upHBV and others[351]
miR-296downHBV and others[206]
miR-487aupHBV and others[269]
miR-638downHBV, HCV and others[352]
miR-940downHBV and others[230]
miR-1246upHBV, HCV[240]
miR-1468upHBV and others[283]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Morishita, A.; Oura, K.; Tadokoro, T.; Fujita, K.; Tani, J.; Masaki, T. MicroRNAs in the Pathogenesis of Hepatocellular Carcinoma: A Review. Cancers 2021, 13, 514. https://doi.org/10.3390/cancers13030514

AMA Style

Morishita A, Oura K, Tadokoro T, Fujita K, Tani J, Masaki T. MicroRNAs in the Pathogenesis of Hepatocellular Carcinoma: A Review. Cancers. 2021; 13(3):514. https://doi.org/10.3390/cancers13030514

Chicago/Turabian Style

Morishita, Asahiro, Kyoko Oura, Tomoko Tadokoro, Koji Fujita, Joji Tani, and Tsutomu Masaki. 2021. "MicroRNAs in the Pathogenesis of Hepatocellular Carcinoma: A Review" Cancers 13, no. 3: 514. https://doi.org/10.3390/cancers13030514

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop